Commentary: MRI Surveillance and Risk Factors in Breast Cancer, April 2024

Article Type
Changed
Mon, 04/01/2024 - 18:05
Dr. Roesch scans the journals, so you don't have to!

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Women with pathogenic BRCA1/2 mutations are presented options of risk-reducing surgery or enhanced surveillance to address their elevated lifetime risk for breast cancer. In regard to breast cancer screening for these women, guidelines recommend annual mammography and breast MRI for those aged 30-75 years; for younger women (age 25-29 years), annual MRI or an individualized schedule on the basis of family history if a breast cancer diagnosis before age 30 is present.[1] Prior studies have highlighted the role of screening MRI in "downstaging," meaning MRI screening detected breast cancers at an earlier stage vs those identified with mammography.[2] As with any screening tool, it is essential to demonstrate the effect of MRI surveillance on mortality for women with BRCA mutations. A cohort study that included 2488 women (age ≥ 30 years) with a BRCA1 (n = 2004) or BRCA2 (n = 484) mutation compared breast cancer mortality rates among those women who participated in MRI screening with those who did not (Lubinski et al). After a median follow-up of 9.2 years, 344 women (13.8%) developed breast cancer, and 35 (1.4%) died from breast cancer. There was an 80% reduction in breast cancer mortality among BRCA1 mutation carriers who participated in MRI surveillance vs those who did not (age-adjusted hazard ratio [HR] 0.20; 95% CI 0.10-0.43; P < .001), but this was not observed for women with BRCA2 mutations (age-adjusted HR 0.87; 95% CI 0.10-17.25; P = .93). At 20 years, the breast cancer mortality rate was 3.2% in the MRI surveillance group compared with 14.9% in the group who did not undergo surveillance. A separate cohort study from Ontario, Canada, including 489 women with BRCA1/2 pathogenic mutations found a 2.0% rate of breast cancer-related mortality at 20 years after the first MRI screening.[3] These data support an intensified surveillance schedule for BRCA mutation carriers, with a need for further research and insight in the BRCA2 population.

A positive family history of cancer and obesity are established risk factors for development of breast cancer among women.[4,5] A population-based cohort study that included 15,055 Chinese women evaluated the association and interaction between body mass index (BMI) and family history of cancer on the risk for breast cancer (Cao et al). The incidence risk for breast cancer was highest in the group with obesity vs the group with normal weight (adjusted HR 2.09; 95% CI 1.42-3.07), and those with a family history of cancer also had an increased risk vs those without a family history of cancer (adjusted HR 1.63; 95% CI 1.22-2.49). Furthermore, women with a BMI ≥ 24 and family history of cancer had a higher risk for breast cancer development compared with women with a BMI < 24 and no family history of cancer (adjusted HR 2.06; 95% CI 1.39-3.06). This study indicates a heightened breast cancer risk when cancer family history and obesity coexist, suggesting the importance of addressing modifiable risk factors and targeting lifestyle interventions in this population.

Triple-negative breast cancer (TNBC), although exhibiting its own heterogeneity, has various features that differentiate this subtype from luminal breast cancers. For example, TNBC generally has a more aggressive course, increased responsiveness to chemotherapy, and earlier pattern of recurrence compared with hormone receptor–positive disease. Prior studies have also shown that established breast cancer risk factors reflect those for the luminal A subtype, whereas those for TNBC are less consistent.[6] A meta-analysis that included 33 studies evaluated the association between traditional breast cancer risk factors and TNBC incidence (Kumar et al). Family history (odds ratio [OR] 1.55; 95% CI 1.34-1.81; P < .001), longer duration of oral contraceptive use (OR 1.29; 95% CI 1.08-1.55; P < .001), and higher breast density (OR 2.19; 95% CI 1.67-2.88; P < .001) were significantly associated with an increased risk for TNBC. Factors including later age at menarche, later age at first birth, and breastfeeding were associated with reduced risk for TNBC. Furthermore, there was no significant association with parity, menopausal hormone therapy, alcohol, smoking, and BMI. This study highlights distinct risk factors that may contribute to higher risk for TNBC, and future research will be valuable to better elucidate the mechanisms at play and to further to understand the differences within this subtype itself.

Additional References

  1. National Comprehensive Cancer Network Clinical Practice Guidelines in Oncology. Genetic/familial high-risk assessment: breast, ovarian, and pancreatic. Version 3.2024. Source 
  2. Saadatmand S, Geuzinge HA, Rutgers EJT, et al; on behalf of the FaMRIsc study group. MRI versus mammography for breast cancer screening in women with familial risk (FaMRIsc): a multicentre, randomised, controlled trial. Lancet Oncol. 2019;20:1136-1147. doi: 10.1016/S1470-2045(19)30275-X  Source
  3. Warner E, Zhu S, Plewes DB, et al. Breast cancer mortality among women with a BRCA1 or BRCA2 mutation in a magnetic resonance imaging plus mammography screening program. Cancers (Basel). 2020;12:3479. doi: 10.3390/cancers12113479 Source
  4. Picon-Ruiz M, Morata-Tarifa C, Valle-Goffin JJ, et al. Obesity and adverse breast cancer risk and outcome: mechanistic insights and strategies for intervention. CA Cancer J Clin. 2017;67:378-397. doi: 10.3322/caac.21405 Source
  5. Engmann NJ, Golmakani MK, Miglioretti DL, et al; for the Breast Cancer Surveillance Consortium. Population-attributable risk proportion of clinical risk factors for breast cancer. JAMA Oncol. 2017;3:1228-1236. doi: 10.1001/jamaoncol.2016.6326 Source
  6. style="font-family:Calibri,sans-serif">Barnard ME, Boeke CE, Tamimi RM. Established breast cancer risk factors and risk of intrinsic tumor subtypes. Biochim Biophys Acta Rev Cancer. 2015;1856:73-85. doi: 10.1016/j.bbcan.2015.0002 Source
Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Publications
Topics
Sections
Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Dr. Roesch scans the journals, so you don't have to!
Dr. Roesch scans the journals, so you don't have to!

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Women with pathogenic BRCA1/2 mutations are presented options of risk-reducing surgery or enhanced surveillance to address their elevated lifetime risk for breast cancer. In regard to breast cancer screening for these women, guidelines recommend annual mammography and breast MRI for those aged 30-75 years; for younger women (age 25-29 years), annual MRI or an individualized schedule on the basis of family history if a breast cancer diagnosis before age 30 is present.[1] Prior studies have highlighted the role of screening MRI in "downstaging," meaning MRI screening detected breast cancers at an earlier stage vs those identified with mammography.[2] As with any screening tool, it is essential to demonstrate the effect of MRI surveillance on mortality for women with BRCA mutations. A cohort study that included 2488 women (age ≥ 30 years) with a BRCA1 (n = 2004) or BRCA2 (n = 484) mutation compared breast cancer mortality rates among those women who participated in MRI screening with those who did not (Lubinski et al). After a median follow-up of 9.2 years, 344 women (13.8%) developed breast cancer, and 35 (1.4%) died from breast cancer. There was an 80% reduction in breast cancer mortality among BRCA1 mutation carriers who participated in MRI surveillance vs those who did not (age-adjusted hazard ratio [HR] 0.20; 95% CI 0.10-0.43; P < .001), but this was not observed for women with BRCA2 mutations (age-adjusted HR 0.87; 95% CI 0.10-17.25; P = .93). At 20 years, the breast cancer mortality rate was 3.2% in the MRI surveillance group compared with 14.9% in the group who did not undergo surveillance. A separate cohort study from Ontario, Canada, including 489 women with BRCA1/2 pathogenic mutations found a 2.0% rate of breast cancer-related mortality at 20 years after the first MRI screening.[3] These data support an intensified surveillance schedule for BRCA mutation carriers, with a need for further research and insight in the BRCA2 population.

A positive family history of cancer and obesity are established risk factors for development of breast cancer among women.[4,5] A population-based cohort study that included 15,055 Chinese women evaluated the association and interaction between body mass index (BMI) and family history of cancer on the risk for breast cancer (Cao et al). The incidence risk for breast cancer was highest in the group with obesity vs the group with normal weight (adjusted HR 2.09; 95% CI 1.42-3.07), and those with a family history of cancer also had an increased risk vs those without a family history of cancer (adjusted HR 1.63; 95% CI 1.22-2.49). Furthermore, women with a BMI ≥ 24 and family history of cancer had a higher risk for breast cancer development compared with women with a BMI < 24 and no family history of cancer (adjusted HR 2.06; 95% CI 1.39-3.06). This study indicates a heightened breast cancer risk when cancer family history and obesity coexist, suggesting the importance of addressing modifiable risk factors and targeting lifestyle interventions in this population.

Triple-negative breast cancer (TNBC), although exhibiting its own heterogeneity, has various features that differentiate this subtype from luminal breast cancers. For example, TNBC generally has a more aggressive course, increased responsiveness to chemotherapy, and earlier pattern of recurrence compared with hormone receptor–positive disease. Prior studies have also shown that established breast cancer risk factors reflect those for the luminal A subtype, whereas those for TNBC are less consistent.[6] A meta-analysis that included 33 studies evaluated the association between traditional breast cancer risk factors and TNBC incidence (Kumar et al). Family history (odds ratio [OR] 1.55; 95% CI 1.34-1.81; P < .001), longer duration of oral contraceptive use (OR 1.29; 95% CI 1.08-1.55; P < .001), and higher breast density (OR 2.19; 95% CI 1.67-2.88; P < .001) were significantly associated with an increased risk for TNBC. Factors including later age at menarche, later age at first birth, and breastfeeding were associated with reduced risk for TNBC. Furthermore, there was no significant association with parity, menopausal hormone therapy, alcohol, smoking, and BMI. This study highlights distinct risk factors that may contribute to higher risk for TNBC, and future research will be valuable to better elucidate the mechanisms at play and to further to understand the differences within this subtype itself.

Additional References

  1. National Comprehensive Cancer Network Clinical Practice Guidelines in Oncology. Genetic/familial high-risk assessment: breast, ovarian, and pancreatic. Version 3.2024. Source 
  2. Saadatmand S, Geuzinge HA, Rutgers EJT, et al; on behalf of the FaMRIsc study group. MRI versus mammography for breast cancer screening in women with familial risk (FaMRIsc): a multicentre, randomised, controlled trial. Lancet Oncol. 2019;20:1136-1147. doi: 10.1016/S1470-2045(19)30275-X  Source
  3. Warner E, Zhu S, Plewes DB, et al. Breast cancer mortality among women with a BRCA1 or BRCA2 mutation in a magnetic resonance imaging plus mammography screening program. Cancers (Basel). 2020;12:3479. doi: 10.3390/cancers12113479 Source
  4. Picon-Ruiz M, Morata-Tarifa C, Valle-Goffin JJ, et al. Obesity and adverse breast cancer risk and outcome: mechanistic insights and strategies for intervention. CA Cancer J Clin. 2017;67:378-397. doi: 10.3322/caac.21405 Source
  5. Engmann NJ, Golmakani MK, Miglioretti DL, et al; for the Breast Cancer Surveillance Consortium. Population-attributable risk proportion of clinical risk factors for breast cancer. JAMA Oncol. 2017;3:1228-1236. doi: 10.1001/jamaoncol.2016.6326 Source
  6. style="font-family:Calibri,sans-serif">Barnard ME, Boeke CE, Tamimi RM. Established breast cancer risk factors and risk of intrinsic tumor subtypes. Biochim Biophys Acta Rev Cancer. 2015;1856:73-85. doi: 10.1016/j.bbcan.2015.0002 Source

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Women with pathogenic BRCA1/2 mutations are presented options of risk-reducing surgery or enhanced surveillance to address their elevated lifetime risk for breast cancer. In regard to breast cancer screening for these women, guidelines recommend annual mammography and breast MRI for those aged 30-75 years; for younger women (age 25-29 years), annual MRI or an individualized schedule on the basis of family history if a breast cancer diagnosis before age 30 is present.[1] Prior studies have highlighted the role of screening MRI in "downstaging," meaning MRI screening detected breast cancers at an earlier stage vs those identified with mammography.[2] As with any screening tool, it is essential to demonstrate the effect of MRI surveillance on mortality for women with BRCA mutations. A cohort study that included 2488 women (age ≥ 30 years) with a BRCA1 (n = 2004) or BRCA2 (n = 484) mutation compared breast cancer mortality rates among those women who participated in MRI screening with those who did not (Lubinski et al). After a median follow-up of 9.2 years, 344 women (13.8%) developed breast cancer, and 35 (1.4%) died from breast cancer. There was an 80% reduction in breast cancer mortality among BRCA1 mutation carriers who participated in MRI surveillance vs those who did not (age-adjusted hazard ratio [HR] 0.20; 95% CI 0.10-0.43; P < .001), but this was not observed for women with BRCA2 mutations (age-adjusted HR 0.87; 95% CI 0.10-17.25; P = .93). At 20 years, the breast cancer mortality rate was 3.2% in the MRI surveillance group compared with 14.9% in the group who did not undergo surveillance. A separate cohort study from Ontario, Canada, including 489 women with BRCA1/2 pathogenic mutations found a 2.0% rate of breast cancer-related mortality at 20 years after the first MRI screening.[3] These data support an intensified surveillance schedule for BRCA mutation carriers, with a need for further research and insight in the BRCA2 population.

A positive family history of cancer and obesity are established risk factors for development of breast cancer among women.[4,5] A population-based cohort study that included 15,055 Chinese women evaluated the association and interaction between body mass index (BMI) and family history of cancer on the risk for breast cancer (Cao et al). The incidence risk for breast cancer was highest in the group with obesity vs the group with normal weight (adjusted HR 2.09; 95% CI 1.42-3.07), and those with a family history of cancer also had an increased risk vs those without a family history of cancer (adjusted HR 1.63; 95% CI 1.22-2.49). Furthermore, women with a BMI ≥ 24 and family history of cancer had a higher risk for breast cancer development compared with women with a BMI < 24 and no family history of cancer (adjusted HR 2.06; 95% CI 1.39-3.06). This study indicates a heightened breast cancer risk when cancer family history and obesity coexist, suggesting the importance of addressing modifiable risk factors and targeting lifestyle interventions in this population.

Triple-negative breast cancer (TNBC), although exhibiting its own heterogeneity, has various features that differentiate this subtype from luminal breast cancers. For example, TNBC generally has a more aggressive course, increased responsiveness to chemotherapy, and earlier pattern of recurrence compared with hormone receptor–positive disease. Prior studies have also shown that established breast cancer risk factors reflect those for the luminal A subtype, whereas those for TNBC are less consistent.[6] A meta-analysis that included 33 studies evaluated the association between traditional breast cancer risk factors and TNBC incidence (Kumar et al). Family history (odds ratio [OR] 1.55; 95% CI 1.34-1.81; P < .001), longer duration of oral contraceptive use (OR 1.29; 95% CI 1.08-1.55; P < .001), and higher breast density (OR 2.19; 95% CI 1.67-2.88; P < .001) were significantly associated with an increased risk for TNBC. Factors including later age at menarche, later age at first birth, and breastfeeding were associated with reduced risk for TNBC. Furthermore, there was no significant association with parity, menopausal hormone therapy, alcohol, smoking, and BMI. This study highlights distinct risk factors that may contribute to higher risk for TNBC, and future research will be valuable to better elucidate the mechanisms at play and to further to understand the differences within this subtype itself.

Additional References

  1. National Comprehensive Cancer Network Clinical Practice Guidelines in Oncology. Genetic/familial high-risk assessment: breast, ovarian, and pancreatic. Version 3.2024. Source 
  2. Saadatmand S, Geuzinge HA, Rutgers EJT, et al; on behalf of the FaMRIsc study group. MRI versus mammography for breast cancer screening in women with familial risk (FaMRIsc): a multicentre, randomised, controlled trial. Lancet Oncol. 2019;20:1136-1147. doi: 10.1016/S1470-2045(19)30275-X  Source
  3. Warner E, Zhu S, Plewes DB, et al. Breast cancer mortality among women with a BRCA1 or BRCA2 mutation in a magnetic resonance imaging plus mammography screening program. Cancers (Basel). 2020;12:3479. doi: 10.3390/cancers12113479 Source
  4. Picon-Ruiz M, Morata-Tarifa C, Valle-Goffin JJ, et al. Obesity and adverse breast cancer risk and outcome: mechanistic insights and strategies for intervention. CA Cancer J Clin. 2017;67:378-397. doi: 10.3322/caac.21405 Source
  5. Engmann NJ, Golmakani MK, Miglioretti DL, et al; for the Breast Cancer Surveillance Consortium. Population-attributable risk proportion of clinical risk factors for breast cancer. JAMA Oncol. 2017;3:1228-1236. doi: 10.1001/jamaoncol.2016.6326 Source
  6. style="font-family:Calibri,sans-serif">Barnard ME, Boeke CE, Tamimi RM. Established breast cancer risk factors and risk of intrinsic tumor subtypes. Biochim Biophys Acta Rev Cancer. 2015;1856:73-85. doi: 10.1016/j.bbcan.2015.0002 Source
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Mon, 05/03/2021 - 14:45
Un-Gate On Date
Mon, 05/03/2021 - 14:45
Use ProPublica
CFC Schedule Remove Status
Mon, 05/03/2021 - 14:45
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Activity Salesforce Deliverable ID
398326.1
Activity ID
109750
Product Name
Clinical Edge Journal Scan
Product ID
124
Supporter Name /ID
Perjeta [ 3532 ]

Commentary: Benign Breast Disease, PD-L1+ TNBC, and Exercise in BC, February 2024

Article Type
Changed
Tue, 02/06/2024 - 09:37
Dr. Roesch scans the journals so you don't have to!

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Benign breast disease (BBD), including nonproliferative disease (NP), proliferative disease without atypia (PDWA), and atypical hyperplasia (AH), is the most common finding after a breast biopsy, representing approximately 75% of cases. Multiple prior studies in the setting of surgical biopsies have shown an association of BBD with an increased risk for breast cancer.[1] Sherman and colleagues investigated this relationship between BBD and breast cancer risk in a contemporary population with use of percutaneous biopsy. This retrospective cohort study included 4819 women with BBD and demonstrated a higher risk in the BBD cohort compared with Surveillance, Epidemiology, and End Results (SEER) data. The study results showed an increase in breast cancer overall (standard incidence ratio (SIR) 1.95; 95% CI 0.176-2.17), invasive breast cancer (SIR 1.56; 95% CI 1.37-1.78), and ductal carcinoma in situ (SIR 3.10; 95% CI 2.54-3.77). Furthermore, increasing BBD severity and an increasing number of foci of BBD were found to have progressively increased the risk. The 10-year breast cancer cumulative incidence was 4.3% for NP, 6.6% for PDWA, and 14.6% for AH compared with an expected cumulative incidence of 2.9% for the general SEER population. This study further helps to inform our understanding of the breast cancer risk associated with BBD and encourages optimization of screening techniques and other diagnostics, modification of lifestyle factors that may influence this risk, and other preventative measures (such as chemoprevention).

The benefit of immunotherapy in combination with chemotherapy for programmed death–ligand 1–positive (PD-L1+) metastatic triple-negative breast cancer (mTNBC) has been shown in both the IMpassion130 and KEYNOTE-355 trials.[2,3] However, the IMpassion131 trial, which evaluated atezolizumab plus paclitaxel, did not show a progression-free survival (PFS) or overall survival (OS) benefit vs paclitaxel alone in PD-L1+ mTNBC.[4] Various explanations for these divergent results have been proposed, including the inherent properties of the chemotherapy backbone, patient populations, and the heterogenous nature of TNBC, which can affect response to immunotherapy. Of present, the various KEYNOTE-355 regimens (pembrolizumab plus investigator's choice chemotherapy [nab-paclitaxel, paclitaxel, or gemcitabine-carboplatin]) are US Food and Drug Administration approved for PD-L1+ mTNBC in the first-line setting. The phase 2 randomized TBCRC 043 trial investigated the effect of atezolizumab with carboplatin in patients with mTNBC and further looked at clinical and molecular correlates of response (Lehmann et al). A total of 106 patients were randomly assigned to carboplatin or carboplatin plus atezolizumab; the combination improved PFS (median PFS, 4.1 vs 2.2 mo; hazard ratio [HR] 0.66; P = .05) and OS (12.6 vs 8.6 mo; HR 0.60; P = .03). Grade 3/4 serious adverse events were more common with carboplatin-atezolizumab vs carboplatin alone (41% vs 8%). In addition, an association of better responses with PD-L1 immunotherapy was seen in patients with obesity, uncontrolled blood glucose levels, high tumor mutation burden, and increased tumor infiltrating lymphocytes. These data support the role of immunotherapy in mTNBC, highlight tumor heterogeneity within this subtype and encourage correlative studies to better define which patients benefit from immunotherapy.

Various studies have demonstrated the favorable impact of physical activity on breast cancer risk in postmenopausal women.[5] However, data in premenopausal women is less clear. Various mechanisms connecting physical activity to premenopausal breast cancer risk have been proposed including the effect of exercise on sex steroid hormones, fasting insulin levels, and inflammation.[6] A pooled analysis from 19 cohort studies including 547,601 premenopausal women, with 10,231 incident cases of breast cancer, aimed to examine the relationship between leisure-time physical activity (sports, exercise, recreational walking) and breast cancer risk in young women (Timmins et al). Higher (90th percentile) vs lower (10th percentile) levels of leisure-time physical activity were associated with a 10% reduction in breast cancer risk after adjustment for body mass index (BMI; adjusted HR 0.90; 95% CI 0.85-0.95; P < .001). They also found a significant reduction in risk: 32% (HR 0.68; P = .01) and 9% (HR 0.91; P = .005) for women with underweight (BMI < 18.5) and with average weight (BMI 18.5-24.9), respectively. Further, the effect of physical activity was most pronounced in the human epidermal growth factor receptor 2 (HER2)–enriched breast cancer subtype, wherein higher vs lower levels of activity were associated with an estimated 45% reduction in breast cancer risk (adjusted HR 0.55; 95% CI 0.37-0.82). These findings support the beneficial role of aerobic exercise and healthy body weight on breast cancer risk among premenopausal women and highlight the value of incorporating this information into counseling for our patients.

Additional References

  1. Figueroa JD, Gierach GL, Duggan MA, et al. Risk factors for breast cancer development by tumor characteristics among women with benign breast disease. Breast Cancer Res. 2021;23:34. doi: 10.1186/s13058-021-01410-1 Source
  2. Schmid P, Adams S, Rugo HS, et al, for the IMpassion130 Trial Investigators. Atezolizumab and nab-paclitaxel in advanced triple-negative breast cancer. N Engl J Med. 2018;379:2108-2121. doi: 10.1056/nejmoa1809615 Source
  3. Cortes J, Rugo HS, Cescon DW, et al, for the KEYNOTE-355 Investigators. Pembrolizumab plus chemotherapy in advanced triple-negative breast cancer. N Engl J Med. 2022;387:217-226. doi: 10.1056/NEJMoa2202809 Source
  4. Miles D, Gligorov J, André F, et al, on behalf of the IMpassion131 investigators. Primary results from IMpassion131, a double-blind, placebo-controlled, randomised phase III trial of first-line paclitaxel with or without atezolizumab for unresectable locally advanced/metastatic triple-negative breast cancer. Ann Oncol. 2021;32:994-1004. doi: 10.1016/j.annonc.2021.05.801 Source
  5. Eliassen AH, Hankinson SE, Rosner B, et al. Physical activity and risk of breast cancer among postmenopausal women. Arch Intern Med. 2010;170:1758-1764. doi: 10.1001/archinternmed.2010.363 Source
  6. Swain CTV, Drummond AE, Boing L, et al. Linking physical activity to breast cancer via sex hormones, part 1: The effect of physical activity on sex steroid hormones. Cancer Epidemiol Biomarkers Prev. 2022;31:16-27. doi: 10.1158/1055-9965.EPI-21-0437 Source
Author and Disclosure Information

Erin Roesch, MD Assistant Professor, Department of Medicine, Cleveland Clinic Lerner College of Medicine; Assocaite Staff Physician, Hematology and Oncology, Cancer Institute, Cleveland, Ohio

Erin E. Roesch, MD, has disclosed the following relevant financial relationships: Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Publications
Topics
Sections
Author and Disclosure Information

Erin Roesch, MD Assistant Professor, Department of Medicine, Cleveland Clinic Lerner College of Medicine; Assocaite Staff Physician, Hematology and Oncology, Cancer Institute, Cleveland, Ohio

Erin E. Roesch, MD, has disclosed the following relevant financial relationships: Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Author and Disclosure Information

Erin Roesch, MD Assistant Professor, Department of Medicine, Cleveland Clinic Lerner College of Medicine; Assocaite Staff Physician, Hematology and Oncology, Cancer Institute, Cleveland, Ohio

Erin E. Roesch, MD, has disclosed the following relevant financial relationships: Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Dr. Roesch scans the journals so you don't have to!
Dr. Roesch scans the journals so you don't have to!

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Benign breast disease (BBD), including nonproliferative disease (NP), proliferative disease without atypia (PDWA), and atypical hyperplasia (AH), is the most common finding after a breast biopsy, representing approximately 75% of cases. Multiple prior studies in the setting of surgical biopsies have shown an association of BBD with an increased risk for breast cancer.[1] Sherman and colleagues investigated this relationship between BBD and breast cancer risk in a contemporary population with use of percutaneous biopsy. This retrospective cohort study included 4819 women with BBD and demonstrated a higher risk in the BBD cohort compared with Surveillance, Epidemiology, and End Results (SEER) data. The study results showed an increase in breast cancer overall (standard incidence ratio (SIR) 1.95; 95% CI 0.176-2.17), invasive breast cancer (SIR 1.56; 95% CI 1.37-1.78), and ductal carcinoma in situ (SIR 3.10; 95% CI 2.54-3.77). Furthermore, increasing BBD severity and an increasing number of foci of BBD were found to have progressively increased the risk. The 10-year breast cancer cumulative incidence was 4.3% for NP, 6.6% for PDWA, and 14.6% for AH compared with an expected cumulative incidence of 2.9% for the general SEER population. This study further helps to inform our understanding of the breast cancer risk associated with BBD and encourages optimization of screening techniques and other diagnostics, modification of lifestyle factors that may influence this risk, and other preventative measures (such as chemoprevention).

The benefit of immunotherapy in combination with chemotherapy for programmed death–ligand 1–positive (PD-L1+) metastatic triple-negative breast cancer (mTNBC) has been shown in both the IMpassion130 and KEYNOTE-355 trials.[2,3] However, the IMpassion131 trial, which evaluated atezolizumab plus paclitaxel, did not show a progression-free survival (PFS) or overall survival (OS) benefit vs paclitaxel alone in PD-L1+ mTNBC.[4] Various explanations for these divergent results have been proposed, including the inherent properties of the chemotherapy backbone, patient populations, and the heterogenous nature of TNBC, which can affect response to immunotherapy. Of present, the various KEYNOTE-355 regimens (pembrolizumab plus investigator's choice chemotherapy [nab-paclitaxel, paclitaxel, or gemcitabine-carboplatin]) are US Food and Drug Administration approved for PD-L1+ mTNBC in the first-line setting. The phase 2 randomized TBCRC 043 trial investigated the effect of atezolizumab with carboplatin in patients with mTNBC and further looked at clinical and molecular correlates of response (Lehmann et al). A total of 106 patients were randomly assigned to carboplatin or carboplatin plus atezolizumab; the combination improved PFS (median PFS, 4.1 vs 2.2 mo; hazard ratio [HR] 0.66; P = .05) and OS (12.6 vs 8.6 mo; HR 0.60; P = .03). Grade 3/4 serious adverse events were more common with carboplatin-atezolizumab vs carboplatin alone (41% vs 8%). In addition, an association of better responses with PD-L1 immunotherapy was seen in patients with obesity, uncontrolled blood glucose levels, high tumor mutation burden, and increased tumor infiltrating lymphocytes. These data support the role of immunotherapy in mTNBC, highlight tumor heterogeneity within this subtype and encourage correlative studies to better define which patients benefit from immunotherapy.

Various studies have demonstrated the favorable impact of physical activity on breast cancer risk in postmenopausal women.[5] However, data in premenopausal women is less clear. Various mechanisms connecting physical activity to premenopausal breast cancer risk have been proposed including the effect of exercise on sex steroid hormones, fasting insulin levels, and inflammation.[6] A pooled analysis from 19 cohort studies including 547,601 premenopausal women, with 10,231 incident cases of breast cancer, aimed to examine the relationship between leisure-time physical activity (sports, exercise, recreational walking) and breast cancer risk in young women (Timmins et al). Higher (90th percentile) vs lower (10th percentile) levels of leisure-time physical activity were associated with a 10% reduction in breast cancer risk after adjustment for body mass index (BMI; adjusted HR 0.90; 95% CI 0.85-0.95; P < .001). They also found a significant reduction in risk: 32% (HR 0.68; P = .01) and 9% (HR 0.91; P = .005) for women with underweight (BMI < 18.5) and with average weight (BMI 18.5-24.9), respectively. Further, the effect of physical activity was most pronounced in the human epidermal growth factor receptor 2 (HER2)–enriched breast cancer subtype, wherein higher vs lower levels of activity were associated with an estimated 45% reduction in breast cancer risk (adjusted HR 0.55; 95% CI 0.37-0.82). These findings support the beneficial role of aerobic exercise and healthy body weight on breast cancer risk among premenopausal women and highlight the value of incorporating this information into counseling for our patients.

Additional References

  1. Figueroa JD, Gierach GL, Duggan MA, et al. Risk factors for breast cancer development by tumor characteristics among women with benign breast disease. Breast Cancer Res. 2021;23:34. doi: 10.1186/s13058-021-01410-1 Source
  2. Schmid P, Adams S, Rugo HS, et al, for the IMpassion130 Trial Investigators. Atezolizumab and nab-paclitaxel in advanced triple-negative breast cancer. N Engl J Med. 2018;379:2108-2121. doi: 10.1056/nejmoa1809615 Source
  3. Cortes J, Rugo HS, Cescon DW, et al, for the KEYNOTE-355 Investigators. Pembrolizumab plus chemotherapy in advanced triple-negative breast cancer. N Engl J Med. 2022;387:217-226. doi: 10.1056/NEJMoa2202809 Source
  4. Miles D, Gligorov J, André F, et al, on behalf of the IMpassion131 investigators. Primary results from IMpassion131, a double-blind, placebo-controlled, randomised phase III trial of first-line paclitaxel with or without atezolizumab for unresectable locally advanced/metastatic triple-negative breast cancer. Ann Oncol. 2021;32:994-1004. doi: 10.1016/j.annonc.2021.05.801 Source
  5. Eliassen AH, Hankinson SE, Rosner B, et al. Physical activity and risk of breast cancer among postmenopausal women. Arch Intern Med. 2010;170:1758-1764. doi: 10.1001/archinternmed.2010.363 Source
  6. Swain CTV, Drummond AE, Boing L, et al. Linking physical activity to breast cancer via sex hormones, part 1: The effect of physical activity on sex steroid hormones. Cancer Epidemiol Biomarkers Prev. 2022;31:16-27. doi: 10.1158/1055-9965.EPI-21-0437 Source

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Benign breast disease (BBD), including nonproliferative disease (NP), proliferative disease without atypia (PDWA), and atypical hyperplasia (AH), is the most common finding after a breast biopsy, representing approximately 75% of cases. Multiple prior studies in the setting of surgical biopsies have shown an association of BBD with an increased risk for breast cancer.[1] Sherman and colleagues investigated this relationship between BBD and breast cancer risk in a contemporary population with use of percutaneous biopsy. This retrospective cohort study included 4819 women with BBD and demonstrated a higher risk in the BBD cohort compared with Surveillance, Epidemiology, and End Results (SEER) data. The study results showed an increase in breast cancer overall (standard incidence ratio (SIR) 1.95; 95% CI 0.176-2.17), invasive breast cancer (SIR 1.56; 95% CI 1.37-1.78), and ductal carcinoma in situ (SIR 3.10; 95% CI 2.54-3.77). Furthermore, increasing BBD severity and an increasing number of foci of BBD were found to have progressively increased the risk. The 10-year breast cancer cumulative incidence was 4.3% for NP, 6.6% for PDWA, and 14.6% for AH compared with an expected cumulative incidence of 2.9% for the general SEER population. This study further helps to inform our understanding of the breast cancer risk associated with BBD and encourages optimization of screening techniques and other diagnostics, modification of lifestyle factors that may influence this risk, and other preventative measures (such as chemoprevention).

The benefit of immunotherapy in combination with chemotherapy for programmed death–ligand 1–positive (PD-L1+) metastatic triple-negative breast cancer (mTNBC) has been shown in both the IMpassion130 and KEYNOTE-355 trials.[2,3] However, the IMpassion131 trial, which evaluated atezolizumab plus paclitaxel, did not show a progression-free survival (PFS) or overall survival (OS) benefit vs paclitaxel alone in PD-L1+ mTNBC.[4] Various explanations for these divergent results have been proposed, including the inherent properties of the chemotherapy backbone, patient populations, and the heterogenous nature of TNBC, which can affect response to immunotherapy. Of present, the various KEYNOTE-355 regimens (pembrolizumab plus investigator's choice chemotherapy [nab-paclitaxel, paclitaxel, or gemcitabine-carboplatin]) are US Food and Drug Administration approved for PD-L1+ mTNBC in the first-line setting. The phase 2 randomized TBCRC 043 trial investigated the effect of atezolizumab with carboplatin in patients with mTNBC and further looked at clinical and molecular correlates of response (Lehmann et al). A total of 106 patients were randomly assigned to carboplatin or carboplatin plus atezolizumab; the combination improved PFS (median PFS, 4.1 vs 2.2 mo; hazard ratio [HR] 0.66; P = .05) and OS (12.6 vs 8.6 mo; HR 0.60; P = .03). Grade 3/4 serious adverse events were more common with carboplatin-atezolizumab vs carboplatin alone (41% vs 8%). In addition, an association of better responses with PD-L1 immunotherapy was seen in patients with obesity, uncontrolled blood glucose levels, high tumor mutation burden, and increased tumor infiltrating lymphocytes. These data support the role of immunotherapy in mTNBC, highlight tumor heterogeneity within this subtype and encourage correlative studies to better define which patients benefit from immunotherapy.

Various studies have demonstrated the favorable impact of physical activity on breast cancer risk in postmenopausal women.[5] However, data in premenopausal women is less clear. Various mechanisms connecting physical activity to premenopausal breast cancer risk have been proposed including the effect of exercise on sex steroid hormones, fasting insulin levels, and inflammation.[6] A pooled analysis from 19 cohort studies including 547,601 premenopausal women, with 10,231 incident cases of breast cancer, aimed to examine the relationship between leisure-time physical activity (sports, exercise, recreational walking) and breast cancer risk in young women (Timmins et al). Higher (90th percentile) vs lower (10th percentile) levels of leisure-time physical activity were associated with a 10% reduction in breast cancer risk after adjustment for body mass index (BMI; adjusted HR 0.90; 95% CI 0.85-0.95; P < .001). They also found a significant reduction in risk: 32% (HR 0.68; P = .01) and 9% (HR 0.91; P = .005) for women with underweight (BMI < 18.5) and with average weight (BMI 18.5-24.9), respectively. Further, the effect of physical activity was most pronounced in the human epidermal growth factor receptor 2 (HER2)–enriched breast cancer subtype, wherein higher vs lower levels of activity were associated with an estimated 45% reduction in breast cancer risk (adjusted HR 0.55; 95% CI 0.37-0.82). These findings support the beneficial role of aerobic exercise and healthy body weight on breast cancer risk among premenopausal women and highlight the value of incorporating this information into counseling for our patients.

Additional References

  1. Figueroa JD, Gierach GL, Duggan MA, et al. Risk factors for breast cancer development by tumor characteristics among women with benign breast disease. Breast Cancer Res. 2021;23:34. doi: 10.1186/s13058-021-01410-1 Source
  2. Schmid P, Adams S, Rugo HS, et al, for the IMpassion130 Trial Investigators. Atezolizumab and nab-paclitaxel in advanced triple-negative breast cancer. N Engl J Med. 2018;379:2108-2121. doi: 10.1056/nejmoa1809615 Source
  3. Cortes J, Rugo HS, Cescon DW, et al, for the KEYNOTE-355 Investigators. Pembrolizumab plus chemotherapy in advanced triple-negative breast cancer. N Engl J Med. 2022;387:217-226. doi: 10.1056/NEJMoa2202809 Source
  4. Miles D, Gligorov J, André F, et al, on behalf of the IMpassion131 investigators. Primary results from IMpassion131, a double-blind, placebo-controlled, randomised phase III trial of first-line paclitaxel with or without atezolizumab for unresectable locally advanced/metastatic triple-negative breast cancer. Ann Oncol. 2021;32:994-1004. doi: 10.1016/j.annonc.2021.05.801 Source
  5. Eliassen AH, Hankinson SE, Rosner B, et al. Physical activity and risk of breast cancer among postmenopausal women. Arch Intern Med. 2010;170:1758-1764. doi: 10.1001/archinternmed.2010.363 Source
  6. Swain CTV, Drummond AE, Boing L, et al. Linking physical activity to breast cancer via sex hormones, part 1: The effect of physical activity on sex steroid hormones. Cancer Epidemiol Biomarkers Prev. 2022;31:16-27. doi: 10.1158/1055-9965.EPI-21-0437 Source
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Fri, 09/29/2023 - 14:00
Un-Gate On Date
Fri, 09/29/2023 - 14:00
Use ProPublica
CFC Schedule Remove Status
Fri, 09/29/2023 - 14:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Activity Salesforce Deliverable ID
398326.1
Activity ID
109750
Product Name
Clinical Edge Journal Scan
Product ID
124
Supporter Name /ID
Perjeta [ 3532 ]

Commentary: Fertility Concerns and Treatment-Related QOL After Breast Cancer, January 2024

Article Type
Changed
Fri, 01/19/2024 - 06:45
Dr. Roesch scans the journals so you don't have to!

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Future fertility is an important consideration for many young women diagnosed with breast cancer, and oncofertility counseling in women with a germline BRCA mutation can carry additional complexities. Examples include concern regarding transmission of the pathogenic gene to offspring, the effect of the deficient gene itself on ovarian function, and the recommendation to pursue risk-reducing bilateral salpingo-oophorectomy at a young age. The safety of pregnancy after breast cancer diagnosis has been demonstrated in several prior studies,1 but data in BRCA carriers have been limited to date. A retrospective cohort study conducted at 78 worldwide centers, including 4732 BRCA carriers diagnosed with breast cancer at age ≤ 40 years, investigated the incidence of pregnancy and maternal and fetal outcomes among these women (Lambertini et al). A total of 659 women had at least one pregnancy each after breast cancer; the cumulative incidence of pregnancy at 10 years was 22% (95% CI 21%-24%), and median time from breast cancer diagnosis to conception was 3.5 years (interquartile range 2.2-5.3 years). There was no significant difference in disease-free survival between those with or without pregnancy after breast cancer (adjusted hazard ratio 0.99; 95% CI 0.81-1.20, P = .90). Continued oncofertility research efforts are essential as the treatment landscape for early-stage breast cancer continues to evolve. Long-term follow-up of the POSITIVE trial included a small group of BRCA carriers and has shown reassuring results regarding interruption of endocrine therapy during attempts at conceiving.2

Young women diagnosed with breast cancer have been shown to experience higher rates of symptoms that may adversely affect quality of life (QOL), including depression, weight gain, vasomotor symptoms, and sexual dysfunction; they may also have a harder time managing these issues.3 Chemotherapy-related amenorrhea (CRA) is one of the side effects of breast cancer treatment that can affect premenopausal women, and is associated with both patient- (age, body mass index) and treatment-related (regimen, duration) factors.4 A study analyzing data derived from the prospective, longitudinal Cancer Toxicities Study included 1636 premenopausal women ≤ 50 years of age with stage I-III breast cancer treated with chemotherapy but not receiving ovarian suppression (Kabirian et al). A total of 83.0% of women reported CRA at year 1, 72.5% at year 2, and 66.1% at year 4. A higher likelihood of CRA was observed for women of older age vs those age 18-34 years (adjusted odds ratio [aOR] for 35-39 years 1.84; 40-44 years 5.90; and ≥ 45 years 21.29; P < .001 for all), those who received adjuvant tamoxifen (aOR 1.97; P < .001), and those who had hot flashes at baseline (aOR 1.83; P = .01). In the QOL analysis, 57.1% reported no recovery of menses. Persistent CRA was associated with worse insomnia, more systemic therapy–related adverse effects, and worse sexual functioning. These findings highlight the importance of identifying and discussing CRA with our patients, as this can have both physical and psychological effects in the survivorship setting.

The phase 3 KEYNOTE-522 trial has established immunotherapy plus an anthracycline-based chemotherapy backbone for the treatment of stage II-III triple-negative breast cancer (TNBC), with improvements in pathologic complete response (pCR) rates and survival outcomes.5 This regimen can present tolerance issues in clinical practice, and rare risks for cardiotoxicity and secondary hematologic malignancies are also relevant to consider. Furthermore, some patients may not be candidates for anthracycline-based treatment due to prior receipt of a drug in this class or cardiac comorbidities. De-escalation strategies are desired to lessen toxicity and maintain (or improve) outcomes. An open-label phase 2 trial (NeoPACT) investigated the efficacy of neoadjuvant carboplatin (AUC 6), docetaxel (75 mg/m2), and pembrolizumab (200 mg) every 21 days for six cycles among 115 patients with stage I-III TNBC (Sharma et al). The overall pCR and residual cancer burden (RCB 0+1) rates were 58% (95% CI 48%-67%) and 69% (95% CI 60%-78%), respectively. Estimated 3-year event-free survival was 86% (95% CI 77%-95%) in all patients, 98% in those with a pCR, and 68% in those with residual disease. This study also demonstrated a positive association of immune biomarkers and pathologic response. The most common grade ≥ 3 treatment-related adverse events were diarrhea (4.3%), anemia (3.5%), and peripheral sensory neuropathy (2.6%). The phase 3 SCARLET (Shorter Anthracycline-Free Chemoimmunotherapy Adapted to Pathologic Response in Early TNBC) trial is comparing the NeoPACT regimen with the standard KEYNOTE-522 regimen in early-stage TNBC and will be critical to further defining this treatment space.6 Presently, considering the described efficacy outcomes with the NeoPACT regimen, this regimen would be very reasonable to consider in patients who are not candidates for an anthracycline. Future prospective evaluation of immune biomarkers and additional predictors of response will also be valuable to further individualize treatment for our patients.

Additional References

  1. Lambertini M, Blondeaux E, Bruzzone M, et al. Pregnancy after breast cancer: A systematic review and meta-analysis. J Clin Oncol. 2021;39:3293-3305. doi: 10.1200/JCO.21.00535
  2. Partridge AH, Niman SM, Ruggeri M, et al, for the International Breast Cancer Study Group and POSITIVE Trial Collaborators. Interrupting endocrine therapy to attempt pregnancy after breast cancer. N Engl J Med. 2023;388:1645-1656. doi: 10.1056/NEJMoa2212856
  3. Howard-Anderson J, Ganz PA, Bower JE, Stanton AL. Quality of life, fertility concerns, and behavioral health outcomes in younger breast cancer survivors: A systematic review. J Natl Cancer Inst. 2012;104:386-405. doi: 10.1093/jnci/djr541
  4. Turnbull AK, Patel S, Martinez-Perez C, et al. Risk of chemotherapy-related amenorrhoea (CRA) in premenopausal women undergoing chemotherapy for early stage breast cancer. Breast Cancer Res Treat. 2021;186:237-245. doi: 10.1007/s10549-020-05951-5
  5. Schmid P, Cortes J, Dent R, et al; KEYNOTE-522 Investigators. Event-free survival with pembrolizumab in early triple-negative breast cancer. N Engl J Med. 2022;386:556-567. doi: 10.1056/NEJMoa2112651
  6. US National Cancer Institute, Cancer Therapy Evaluation Program. Shorter anthracycline-free chemoimmunotherapy adapted to pathological response in early TNBC (SCARLET); SWOG S2212. Source
Author and Disclosure Information

Erin Roesch, MD Assistant Professor, Department of Medicine, Cleveland Clinic Lerner College of Medicine; Assocaite Staff Physician, Hematology and Oncology, Cancer Institute, Cleveland, Ohio

Erin E. Roesch, MD, has disclosed the following relevant financial relationships: Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Publications
Topics
Sections
Author and Disclosure Information

Erin Roesch, MD Assistant Professor, Department of Medicine, Cleveland Clinic Lerner College of Medicine; Assocaite Staff Physician, Hematology and Oncology, Cancer Institute, Cleveland, Ohio

Erin E. Roesch, MD, has disclosed the following relevant financial relationships: Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Author and Disclosure Information

Erin Roesch, MD Assistant Professor, Department of Medicine, Cleveland Clinic Lerner College of Medicine; Assocaite Staff Physician, Hematology and Oncology, Cancer Institute, Cleveland, Ohio

Erin E. Roesch, MD, has disclosed the following relevant financial relationships: Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Dr. Roesch scans the journals so you don't have to!
Dr. Roesch scans the journals so you don't have to!

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Future fertility is an important consideration for many young women diagnosed with breast cancer, and oncofertility counseling in women with a germline BRCA mutation can carry additional complexities. Examples include concern regarding transmission of the pathogenic gene to offspring, the effect of the deficient gene itself on ovarian function, and the recommendation to pursue risk-reducing bilateral salpingo-oophorectomy at a young age. The safety of pregnancy after breast cancer diagnosis has been demonstrated in several prior studies,1 but data in BRCA carriers have been limited to date. A retrospective cohort study conducted at 78 worldwide centers, including 4732 BRCA carriers diagnosed with breast cancer at age ≤ 40 years, investigated the incidence of pregnancy and maternal and fetal outcomes among these women (Lambertini et al). A total of 659 women had at least one pregnancy each after breast cancer; the cumulative incidence of pregnancy at 10 years was 22% (95% CI 21%-24%), and median time from breast cancer diagnosis to conception was 3.5 years (interquartile range 2.2-5.3 years). There was no significant difference in disease-free survival between those with or without pregnancy after breast cancer (adjusted hazard ratio 0.99; 95% CI 0.81-1.20, P = .90). Continued oncofertility research efforts are essential as the treatment landscape for early-stage breast cancer continues to evolve. Long-term follow-up of the POSITIVE trial included a small group of BRCA carriers and has shown reassuring results regarding interruption of endocrine therapy during attempts at conceiving.2

Young women diagnosed with breast cancer have been shown to experience higher rates of symptoms that may adversely affect quality of life (QOL), including depression, weight gain, vasomotor symptoms, and sexual dysfunction; they may also have a harder time managing these issues.3 Chemotherapy-related amenorrhea (CRA) is one of the side effects of breast cancer treatment that can affect premenopausal women, and is associated with both patient- (age, body mass index) and treatment-related (regimen, duration) factors.4 A study analyzing data derived from the prospective, longitudinal Cancer Toxicities Study included 1636 premenopausal women ≤ 50 years of age with stage I-III breast cancer treated with chemotherapy but not receiving ovarian suppression (Kabirian et al). A total of 83.0% of women reported CRA at year 1, 72.5% at year 2, and 66.1% at year 4. A higher likelihood of CRA was observed for women of older age vs those age 18-34 years (adjusted odds ratio [aOR] for 35-39 years 1.84; 40-44 years 5.90; and ≥ 45 years 21.29; P < .001 for all), those who received adjuvant tamoxifen (aOR 1.97; P < .001), and those who had hot flashes at baseline (aOR 1.83; P = .01). In the QOL analysis, 57.1% reported no recovery of menses. Persistent CRA was associated with worse insomnia, more systemic therapy–related adverse effects, and worse sexual functioning. These findings highlight the importance of identifying and discussing CRA with our patients, as this can have both physical and psychological effects in the survivorship setting.

The phase 3 KEYNOTE-522 trial has established immunotherapy plus an anthracycline-based chemotherapy backbone for the treatment of stage II-III triple-negative breast cancer (TNBC), with improvements in pathologic complete response (pCR) rates and survival outcomes.5 This regimen can present tolerance issues in clinical practice, and rare risks for cardiotoxicity and secondary hematologic malignancies are also relevant to consider. Furthermore, some patients may not be candidates for anthracycline-based treatment due to prior receipt of a drug in this class or cardiac comorbidities. De-escalation strategies are desired to lessen toxicity and maintain (or improve) outcomes. An open-label phase 2 trial (NeoPACT) investigated the efficacy of neoadjuvant carboplatin (AUC 6), docetaxel (75 mg/m2), and pembrolizumab (200 mg) every 21 days for six cycles among 115 patients with stage I-III TNBC (Sharma et al). The overall pCR and residual cancer burden (RCB 0+1) rates were 58% (95% CI 48%-67%) and 69% (95% CI 60%-78%), respectively. Estimated 3-year event-free survival was 86% (95% CI 77%-95%) in all patients, 98% in those with a pCR, and 68% in those with residual disease. This study also demonstrated a positive association of immune biomarkers and pathologic response. The most common grade ≥ 3 treatment-related adverse events were diarrhea (4.3%), anemia (3.5%), and peripheral sensory neuropathy (2.6%). The phase 3 SCARLET (Shorter Anthracycline-Free Chemoimmunotherapy Adapted to Pathologic Response in Early TNBC) trial is comparing the NeoPACT regimen with the standard KEYNOTE-522 regimen in early-stage TNBC and will be critical to further defining this treatment space.6 Presently, considering the described efficacy outcomes with the NeoPACT regimen, this regimen would be very reasonable to consider in patients who are not candidates for an anthracycline. Future prospective evaluation of immune biomarkers and additional predictors of response will also be valuable to further individualize treatment for our patients.

Additional References

  1. Lambertini M, Blondeaux E, Bruzzone M, et al. Pregnancy after breast cancer: A systematic review and meta-analysis. J Clin Oncol. 2021;39:3293-3305. doi: 10.1200/JCO.21.00535
  2. Partridge AH, Niman SM, Ruggeri M, et al, for the International Breast Cancer Study Group and POSITIVE Trial Collaborators. Interrupting endocrine therapy to attempt pregnancy after breast cancer. N Engl J Med. 2023;388:1645-1656. doi: 10.1056/NEJMoa2212856
  3. Howard-Anderson J, Ganz PA, Bower JE, Stanton AL. Quality of life, fertility concerns, and behavioral health outcomes in younger breast cancer survivors: A systematic review. J Natl Cancer Inst. 2012;104:386-405. doi: 10.1093/jnci/djr541
  4. Turnbull AK, Patel S, Martinez-Perez C, et al. Risk of chemotherapy-related amenorrhoea (CRA) in premenopausal women undergoing chemotherapy for early stage breast cancer. Breast Cancer Res Treat. 2021;186:237-245. doi: 10.1007/s10549-020-05951-5
  5. Schmid P, Cortes J, Dent R, et al; KEYNOTE-522 Investigators. Event-free survival with pembrolizumab in early triple-negative breast cancer. N Engl J Med. 2022;386:556-567. doi: 10.1056/NEJMoa2112651
  6. US National Cancer Institute, Cancer Therapy Evaluation Program. Shorter anthracycline-free chemoimmunotherapy adapted to pathological response in early TNBC (SCARLET); SWOG S2212. Source

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Future fertility is an important consideration for many young women diagnosed with breast cancer, and oncofertility counseling in women with a germline BRCA mutation can carry additional complexities. Examples include concern regarding transmission of the pathogenic gene to offspring, the effect of the deficient gene itself on ovarian function, and the recommendation to pursue risk-reducing bilateral salpingo-oophorectomy at a young age. The safety of pregnancy after breast cancer diagnosis has been demonstrated in several prior studies,1 but data in BRCA carriers have been limited to date. A retrospective cohort study conducted at 78 worldwide centers, including 4732 BRCA carriers diagnosed with breast cancer at age ≤ 40 years, investigated the incidence of pregnancy and maternal and fetal outcomes among these women (Lambertini et al). A total of 659 women had at least one pregnancy each after breast cancer; the cumulative incidence of pregnancy at 10 years was 22% (95% CI 21%-24%), and median time from breast cancer diagnosis to conception was 3.5 years (interquartile range 2.2-5.3 years). There was no significant difference in disease-free survival between those with or without pregnancy after breast cancer (adjusted hazard ratio 0.99; 95% CI 0.81-1.20, P = .90). Continued oncofertility research efforts are essential as the treatment landscape for early-stage breast cancer continues to evolve. Long-term follow-up of the POSITIVE trial included a small group of BRCA carriers and has shown reassuring results regarding interruption of endocrine therapy during attempts at conceiving.2

Young women diagnosed with breast cancer have been shown to experience higher rates of symptoms that may adversely affect quality of life (QOL), including depression, weight gain, vasomotor symptoms, and sexual dysfunction; they may also have a harder time managing these issues.3 Chemotherapy-related amenorrhea (CRA) is one of the side effects of breast cancer treatment that can affect premenopausal women, and is associated with both patient- (age, body mass index) and treatment-related (regimen, duration) factors.4 A study analyzing data derived from the prospective, longitudinal Cancer Toxicities Study included 1636 premenopausal women ≤ 50 years of age with stage I-III breast cancer treated with chemotherapy but not receiving ovarian suppression (Kabirian et al). A total of 83.0% of women reported CRA at year 1, 72.5% at year 2, and 66.1% at year 4. A higher likelihood of CRA was observed for women of older age vs those age 18-34 years (adjusted odds ratio [aOR] for 35-39 years 1.84; 40-44 years 5.90; and ≥ 45 years 21.29; P < .001 for all), those who received adjuvant tamoxifen (aOR 1.97; P < .001), and those who had hot flashes at baseline (aOR 1.83; P = .01). In the QOL analysis, 57.1% reported no recovery of menses. Persistent CRA was associated with worse insomnia, more systemic therapy–related adverse effects, and worse sexual functioning. These findings highlight the importance of identifying and discussing CRA with our patients, as this can have both physical and psychological effects in the survivorship setting.

The phase 3 KEYNOTE-522 trial has established immunotherapy plus an anthracycline-based chemotherapy backbone for the treatment of stage II-III triple-negative breast cancer (TNBC), with improvements in pathologic complete response (pCR) rates and survival outcomes.5 This regimen can present tolerance issues in clinical practice, and rare risks for cardiotoxicity and secondary hematologic malignancies are also relevant to consider. Furthermore, some patients may not be candidates for anthracycline-based treatment due to prior receipt of a drug in this class or cardiac comorbidities. De-escalation strategies are desired to lessen toxicity and maintain (or improve) outcomes. An open-label phase 2 trial (NeoPACT) investigated the efficacy of neoadjuvant carboplatin (AUC 6), docetaxel (75 mg/m2), and pembrolizumab (200 mg) every 21 days for six cycles among 115 patients with stage I-III TNBC (Sharma et al). The overall pCR and residual cancer burden (RCB 0+1) rates were 58% (95% CI 48%-67%) and 69% (95% CI 60%-78%), respectively. Estimated 3-year event-free survival was 86% (95% CI 77%-95%) in all patients, 98% in those with a pCR, and 68% in those with residual disease. This study also demonstrated a positive association of immune biomarkers and pathologic response. The most common grade ≥ 3 treatment-related adverse events were diarrhea (4.3%), anemia (3.5%), and peripheral sensory neuropathy (2.6%). The phase 3 SCARLET (Shorter Anthracycline-Free Chemoimmunotherapy Adapted to Pathologic Response in Early TNBC) trial is comparing the NeoPACT regimen with the standard KEYNOTE-522 regimen in early-stage TNBC and will be critical to further defining this treatment space.6 Presently, considering the described efficacy outcomes with the NeoPACT regimen, this regimen would be very reasonable to consider in patients who are not candidates for an anthracycline. Future prospective evaluation of immune biomarkers and additional predictors of response will also be valuable to further individualize treatment for our patients.

Additional References

  1. Lambertini M, Blondeaux E, Bruzzone M, et al. Pregnancy after breast cancer: A systematic review and meta-analysis. J Clin Oncol. 2021;39:3293-3305. doi: 10.1200/JCO.21.00535
  2. Partridge AH, Niman SM, Ruggeri M, et al, for the International Breast Cancer Study Group and POSITIVE Trial Collaborators. Interrupting endocrine therapy to attempt pregnancy after breast cancer. N Engl J Med. 2023;388:1645-1656. doi: 10.1056/NEJMoa2212856
  3. Howard-Anderson J, Ganz PA, Bower JE, Stanton AL. Quality of life, fertility concerns, and behavioral health outcomes in younger breast cancer survivors: A systematic review. J Natl Cancer Inst. 2012;104:386-405. doi: 10.1093/jnci/djr541
  4. Turnbull AK, Patel S, Martinez-Perez C, et al. Risk of chemotherapy-related amenorrhoea (CRA) in premenopausal women undergoing chemotherapy for early stage breast cancer. Breast Cancer Res Treat. 2021;186:237-245. doi: 10.1007/s10549-020-05951-5
  5. Schmid P, Cortes J, Dent R, et al; KEYNOTE-522 Investigators. Event-free survival with pembrolizumab in early triple-negative breast cancer. N Engl J Med. 2022;386:556-567. doi: 10.1056/NEJMoa2112651
  6. US National Cancer Institute, Cancer Therapy Evaluation Program. Shorter anthracycline-free chemoimmunotherapy adapted to pathological response in early TNBC (SCARLET); SWOG S2212. Source
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Fri, 09/29/2023 - 14:00
Un-Gate On Date
Fri, 09/29/2023 - 14:00
Use ProPublica
CFC Schedule Remove Status
Fri, 09/29/2023 - 14:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Activity Salesforce Deliverable ID
398326.1
Activity ID
109750
Product Name
Clinical Edge Journal Scan
Product ID
124
Supporter Name /ID
Perjeta [ 3532 ]

Commentary: Obesity, Pregnancy, and Adjuvant Chemotherapy in BC, December 2023

Article Type
Changed
Wed, 12/20/2023 - 18:08
Dr. Roesch scans the journals so you don't have to!

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Obesity and other metabolic comorbidities (including glucose intolerance, hypertension, and dyslipidemia) have been associated with poorer prognosis among breast cancer patients.1 The underlying mechanisms for which obesity is linked to inferior breast cancer outcomes is complex and may also involve drug efficacy in these patients. Data from the ATAC trial showed that there was a higher risk for recurrence among patients with obesity who were receiving an aromatase inhibitor (AI) vs patients with a healthy body weight receiving an AI; however, patients receiving tamoxifen did not exhibit this difference.2 A Danish Breast Cancer Group cohort study including 13,230 postmenopausal patients with stage I-III hormone receptor–positive (HR+) breast cancer treated with AI investigated the association of body mass index with recurrence (Harborg et al). There was a significantly increased risk for recurrence among those patients with obesity (adjusted hazard ratio 1.18; 95% CI 1.01-1.37) and severe obesity (adjusted hazard ratio 1.32; 95% CI 1.08-1.62) vs patients with healthy body weight. These results highlight the importance of lifestyle interventions targeting obesity and metabolic factors in breast cancer patients and support future studies investigating optimal drug selection based on body composition.

Breast cancer in young women presents a unique set of challenges owing to life-stage at the time of diagnosis and treatment. Oncofertility, family planning, and pregnancy are essential issues to address at the time of initial consultation and throughout the survivorship setting. Various studies have provided supportive evidence regarding the safety of pregnancy after breast cancer diagnosis and treatment.3 HR+ breast cancer is associated with its own distinctive considerations related to pregnancy and its timing, including the use of endocrine therapy for 5-10 years, the role of female hormones during pregnancy, and late patterns of recurrence that characterize this subtype. A meta-analysis including eight eligible studies and 3805 women with HR+ early breast cancer investigated the prognostic impact of future pregnancy among these patients (Arecco et al). A total of 1285 women had a pregnancy after breast cancer diagnosis and treatment; there was no difference in disease-free survival (hazard ratio 0.96; 95% CI 0.75-1.24; P = .781) and better overall survival (OS; hazard ratio 0.46; 95% CI 0.27-0.77; P < .005) in those with vs those without subsequent pregnancy. Added to this body of data is the prospective POSITIVE trial, which showed that a temporary pause of endocrine therapy for an attempt at conceiving appears to be safe in young women with early HR+ breast cancer with short-term follow-up.4 Future research efforts investigating outcomes after assisted reproductive technologies in this population, those with germline mutations, and extended follow-up of studies, such as POSITIVE, will continue to inform guidance for and management of young women with breast cancer.

Guidelines favor the use of adjuvant chemotherapy for small, node-negative, triple-negative breast cancer (TNBC), specifically T1b and T1c tumors.5 However, high-quality data to inform this decision-making are sparse, and it is valuable to consider the magnitude of benefit weighed against possible risks and side effects of treatment, as well as patient comorbidities. A retrospective analysis of the Surveillance, Epidemiology, and End Results (SEER) database including 11,510 patients (3388 with T1b and 8122 with T1c TNBC) evaluated the impact of adjuvant chemotherapy on OS and breast cancer–specific survival (BCSS) (Carbajal-Ochoa et al). The use of adjuvant chemotherapy was associated with improved OS (hazard ratio 0.54; 95% CI 0.47-0.62; P < .001) and BCSS (hazard ratio 0.79; 95% CI 0.63-0.99; P = .043) among T1c TNBC. For those with T1b tumors, adjuvant chemotherapy improved OS (hazard ratio 0.52; 95% CI 0.41-0.68; P < .001) but did not improve BCSS (hazard ratio 0.70; 95% CI 0.45-1.07; P = .10). A better understanding of the molecular drivers implicated in this heterogeneous subtype, and predictors of response and resistance, will aid in identifying those patients who have greater benefit and those who can potentially be spared chemotherapy-related toxicities.

Additional References

  1. Anwar SL, Cahyono R, Prabowo D, et al. Metabolic comorbidities and the association with risks of recurrent metastatic disease in breast cancer survivors. BMC Cancer. 2021;21:590. doi: 10.1186/s12885-021-08343-0>
  2. Sestak I, Distler W, Forbes JF, et al. Effect of body mass index on recurrences in tamoxifen and anastrozole treated women: An exploratory analysis from the ATAC trial. J Clin Oncol. 2010;28:3411-3415. doi: 10.1200/JCO.2009.27.2021
  3. Lambertini M, Blondeaux E, Bruzzone M, et al. Pregnancy after breast cancer: A systematic review and meta-analysis. J Clin Oncol. 2021;39:3293-3305. doi: 10.1200/JCO.21.00535
  4. Partridge AH, Niman SM, Ruggeri M, et al for the International Breast Cancer Study Group and POSITIVE Trial Collaborators. Interrupting endocrine therapy to attempt pregnancy after breast cancer. N Engl J Med. 2023;388:1645-1656. doi: 10.1056/NEJMoa2212856
  5. Curigliano G, Burstein HJ, Winer EP, et al. De-escalating and escalating treatments for early-stage breast cancer: The St. Gallen International Expert Consensus Conference on the Primary Therapy of Early Breast Cancer 2017. Ann Oncol. 2017;28:1700-1712. doi: 10.1093/annonc/mdx308
Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Publications
Topics
Sections
Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Dr. Roesch scans the journals so you don't have to!
Dr. Roesch scans the journals so you don't have to!

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Obesity and other metabolic comorbidities (including glucose intolerance, hypertension, and dyslipidemia) have been associated with poorer prognosis among breast cancer patients.1 The underlying mechanisms for which obesity is linked to inferior breast cancer outcomes is complex and may also involve drug efficacy in these patients. Data from the ATAC trial showed that there was a higher risk for recurrence among patients with obesity who were receiving an aromatase inhibitor (AI) vs patients with a healthy body weight receiving an AI; however, patients receiving tamoxifen did not exhibit this difference.2 A Danish Breast Cancer Group cohort study including 13,230 postmenopausal patients with stage I-III hormone receptor–positive (HR+) breast cancer treated with AI investigated the association of body mass index with recurrence (Harborg et al). There was a significantly increased risk for recurrence among those patients with obesity (adjusted hazard ratio 1.18; 95% CI 1.01-1.37) and severe obesity (adjusted hazard ratio 1.32; 95% CI 1.08-1.62) vs patients with healthy body weight. These results highlight the importance of lifestyle interventions targeting obesity and metabolic factors in breast cancer patients and support future studies investigating optimal drug selection based on body composition.

Breast cancer in young women presents a unique set of challenges owing to life-stage at the time of diagnosis and treatment. Oncofertility, family planning, and pregnancy are essential issues to address at the time of initial consultation and throughout the survivorship setting. Various studies have provided supportive evidence regarding the safety of pregnancy after breast cancer diagnosis and treatment.3 HR+ breast cancer is associated with its own distinctive considerations related to pregnancy and its timing, including the use of endocrine therapy for 5-10 years, the role of female hormones during pregnancy, and late patterns of recurrence that characterize this subtype. A meta-analysis including eight eligible studies and 3805 women with HR+ early breast cancer investigated the prognostic impact of future pregnancy among these patients (Arecco et al). A total of 1285 women had a pregnancy after breast cancer diagnosis and treatment; there was no difference in disease-free survival (hazard ratio 0.96; 95% CI 0.75-1.24; P = .781) and better overall survival (OS; hazard ratio 0.46; 95% CI 0.27-0.77; P < .005) in those with vs those without subsequent pregnancy. Added to this body of data is the prospective POSITIVE trial, which showed that a temporary pause of endocrine therapy for an attempt at conceiving appears to be safe in young women with early HR+ breast cancer with short-term follow-up.4 Future research efforts investigating outcomes after assisted reproductive technologies in this population, those with germline mutations, and extended follow-up of studies, such as POSITIVE, will continue to inform guidance for and management of young women with breast cancer.

Guidelines favor the use of adjuvant chemotherapy for small, node-negative, triple-negative breast cancer (TNBC), specifically T1b and T1c tumors.5 However, high-quality data to inform this decision-making are sparse, and it is valuable to consider the magnitude of benefit weighed against possible risks and side effects of treatment, as well as patient comorbidities. A retrospective analysis of the Surveillance, Epidemiology, and End Results (SEER) database including 11,510 patients (3388 with T1b and 8122 with T1c TNBC) evaluated the impact of adjuvant chemotherapy on OS and breast cancer–specific survival (BCSS) (Carbajal-Ochoa et al). The use of adjuvant chemotherapy was associated with improved OS (hazard ratio 0.54; 95% CI 0.47-0.62; P < .001) and BCSS (hazard ratio 0.79; 95% CI 0.63-0.99; P = .043) among T1c TNBC. For those with T1b tumors, adjuvant chemotherapy improved OS (hazard ratio 0.52; 95% CI 0.41-0.68; P < .001) but did not improve BCSS (hazard ratio 0.70; 95% CI 0.45-1.07; P = .10). A better understanding of the molecular drivers implicated in this heterogeneous subtype, and predictors of response and resistance, will aid in identifying those patients who have greater benefit and those who can potentially be spared chemotherapy-related toxicities.

Additional References

  1. Anwar SL, Cahyono R, Prabowo D, et al. Metabolic comorbidities and the association with risks of recurrent metastatic disease in breast cancer survivors. BMC Cancer. 2021;21:590. doi: 10.1186/s12885-021-08343-0>
  2. Sestak I, Distler W, Forbes JF, et al. Effect of body mass index on recurrences in tamoxifen and anastrozole treated women: An exploratory analysis from the ATAC trial. J Clin Oncol. 2010;28:3411-3415. doi: 10.1200/JCO.2009.27.2021
  3. Lambertini M, Blondeaux E, Bruzzone M, et al. Pregnancy after breast cancer: A systematic review and meta-analysis. J Clin Oncol. 2021;39:3293-3305. doi: 10.1200/JCO.21.00535
  4. Partridge AH, Niman SM, Ruggeri M, et al for the International Breast Cancer Study Group and POSITIVE Trial Collaborators. Interrupting endocrine therapy to attempt pregnancy after breast cancer. N Engl J Med. 2023;388:1645-1656. doi: 10.1056/NEJMoa2212856
  5. Curigliano G, Burstein HJ, Winer EP, et al. De-escalating and escalating treatments for early-stage breast cancer: The St. Gallen International Expert Consensus Conference on the Primary Therapy of Early Breast Cancer 2017. Ann Oncol. 2017;28:1700-1712. doi: 10.1093/annonc/mdx308

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Obesity and other metabolic comorbidities (including glucose intolerance, hypertension, and dyslipidemia) have been associated with poorer prognosis among breast cancer patients.1 The underlying mechanisms for which obesity is linked to inferior breast cancer outcomes is complex and may also involve drug efficacy in these patients. Data from the ATAC trial showed that there was a higher risk for recurrence among patients with obesity who were receiving an aromatase inhibitor (AI) vs patients with a healthy body weight receiving an AI; however, patients receiving tamoxifen did not exhibit this difference.2 A Danish Breast Cancer Group cohort study including 13,230 postmenopausal patients with stage I-III hormone receptor–positive (HR+) breast cancer treated with AI investigated the association of body mass index with recurrence (Harborg et al). There was a significantly increased risk for recurrence among those patients with obesity (adjusted hazard ratio 1.18; 95% CI 1.01-1.37) and severe obesity (adjusted hazard ratio 1.32; 95% CI 1.08-1.62) vs patients with healthy body weight. These results highlight the importance of lifestyle interventions targeting obesity and metabolic factors in breast cancer patients and support future studies investigating optimal drug selection based on body composition.

Breast cancer in young women presents a unique set of challenges owing to life-stage at the time of diagnosis and treatment. Oncofertility, family planning, and pregnancy are essential issues to address at the time of initial consultation and throughout the survivorship setting. Various studies have provided supportive evidence regarding the safety of pregnancy after breast cancer diagnosis and treatment.3 HR+ breast cancer is associated with its own distinctive considerations related to pregnancy and its timing, including the use of endocrine therapy for 5-10 years, the role of female hormones during pregnancy, and late patterns of recurrence that characterize this subtype. A meta-analysis including eight eligible studies and 3805 women with HR+ early breast cancer investigated the prognostic impact of future pregnancy among these patients (Arecco et al). A total of 1285 women had a pregnancy after breast cancer diagnosis and treatment; there was no difference in disease-free survival (hazard ratio 0.96; 95% CI 0.75-1.24; P = .781) and better overall survival (OS; hazard ratio 0.46; 95% CI 0.27-0.77; P < .005) in those with vs those without subsequent pregnancy. Added to this body of data is the prospective POSITIVE trial, which showed that a temporary pause of endocrine therapy for an attempt at conceiving appears to be safe in young women with early HR+ breast cancer with short-term follow-up.4 Future research efforts investigating outcomes after assisted reproductive technologies in this population, those with germline mutations, and extended follow-up of studies, such as POSITIVE, will continue to inform guidance for and management of young women with breast cancer.

Guidelines favor the use of adjuvant chemotherapy for small, node-negative, triple-negative breast cancer (TNBC), specifically T1b and T1c tumors.5 However, high-quality data to inform this decision-making are sparse, and it is valuable to consider the magnitude of benefit weighed against possible risks and side effects of treatment, as well as patient comorbidities. A retrospective analysis of the Surveillance, Epidemiology, and End Results (SEER) database including 11,510 patients (3388 with T1b and 8122 with T1c TNBC) evaluated the impact of adjuvant chemotherapy on OS and breast cancer–specific survival (BCSS) (Carbajal-Ochoa et al). The use of adjuvant chemotherapy was associated with improved OS (hazard ratio 0.54; 95% CI 0.47-0.62; P < .001) and BCSS (hazard ratio 0.79; 95% CI 0.63-0.99; P = .043) among T1c TNBC. For those with T1b tumors, adjuvant chemotherapy improved OS (hazard ratio 0.52; 95% CI 0.41-0.68; P < .001) but did not improve BCSS (hazard ratio 0.70; 95% CI 0.45-1.07; P = .10). A better understanding of the molecular drivers implicated in this heterogeneous subtype, and predictors of response and resistance, will aid in identifying those patients who have greater benefit and those who can potentially be spared chemotherapy-related toxicities.

Additional References

  1. Anwar SL, Cahyono R, Prabowo D, et al. Metabolic comorbidities and the association with risks of recurrent metastatic disease in breast cancer survivors. BMC Cancer. 2021;21:590. doi: 10.1186/s12885-021-08343-0>
  2. Sestak I, Distler W, Forbes JF, et al. Effect of body mass index on recurrences in tamoxifen and anastrozole treated women: An exploratory analysis from the ATAC trial. J Clin Oncol. 2010;28:3411-3415. doi: 10.1200/JCO.2009.27.2021
  3. Lambertini M, Blondeaux E, Bruzzone M, et al. Pregnancy after breast cancer: A systematic review and meta-analysis. J Clin Oncol. 2021;39:3293-3305. doi: 10.1200/JCO.21.00535
  4. Partridge AH, Niman SM, Ruggeri M, et al for the International Breast Cancer Study Group and POSITIVE Trial Collaborators. Interrupting endocrine therapy to attempt pregnancy after breast cancer. N Engl J Med. 2023;388:1645-1656. doi: 10.1056/NEJMoa2212856
  5. Curigliano G, Burstein HJ, Winer EP, et al. De-escalating and escalating treatments for early-stage breast cancer: The St. Gallen International Expert Consensus Conference on the Primary Therapy of Early Breast Cancer 2017. Ann Oncol. 2017;28:1700-1712. doi: 10.1093/annonc/mdx308
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Fri, 09/29/2023 - 14:00
Un-Gate On Date
Fri, 09/29/2023 - 14:00
Use ProPublica
CFC Schedule Remove Status
Fri, 09/29/2023 - 14:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Activity Salesforce Deliverable ID
367005.2
Activity ID
93656
Product Name
Clinical Edge Journal Scan
Product ID
124
Supporter Name /ID
Perjeta [ 3532 ]

Commentary: Obesity, Pregnancy, and Adjuvant Chemotherapy in BC, December 2023

Article Type
Changed
Tue, 11/21/2023 - 15:48
Dr. Roesch scans the journals so you don't have to!

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Obesity and other metabolic comorbidities (including glucose intolerance, hypertension, and dyslipidemia) have been associated with poorer prognosis among breast cancer patients.1 The underlying mechanisms for which obesity is linked to inferior breast cancer outcomes is complex and may also involve drug efficacy in these patients. Data from the ATAC trial showed that there was a higher risk for recurrence among patients with obesity who were receiving an aromatase inhibitor (AI) vs patients with a healthy body weight receiving an AI; however, patients receiving tamoxifen did not exhibit this difference.2 A Danish Breast Cancer Group cohort study including 13,230 postmenopausal patients with stage I-III hormone receptor–positive (HR+) breast cancer treated with AI investigated the association of body mass index with recurrence (Harborg et al). There was a significantly increased risk for recurrence among those patients with obesity (adjusted hazard ratio 1.18; 95% CI 1.01-1.37) and severe obesity (adjusted hazard ratio 1.32; 95% CI 1.08-1.62) vs patients with healthy body weight. These results highlight the importance of lifestyle interventions targeting obesity and metabolic factors in breast cancer patients and support future studies investigating optimal drug selection based on body composition.

Breast cancer in young women presents a unique set of challenges owing to life-stage at the time of diagnosis and treatment. Oncofertility, family planning, and pregnancy are essential issues to address at the time of initial consultation and throughout the survivorship setting. Various studies have provided supportive evidence regarding the safety of pregnancy after breast cancer diagnosis and treatment.3 HR+ breast cancer is associated with its own distinctive considerations related to pregnancy and its timing, including the use of endocrine therapy for 5-10 years, the role of female hormones during pregnancy, and late patterns of recurrence that characterize this subtype. A meta-analysis including eight eligible studies and 3805 women with HR+ early breast cancer investigated the prognostic impact of future pregnancy among these patients (Arecco et al). A total of 1285 women had a pregnancy after breast cancer diagnosis and treatment; there was no difference in disease-free survival (hazard ratio 0.96; 95% CI 0.75-1.24; P = .781) and better overall survival (OS; hazard ratio 0.46; 95% CI 0.27-0.77; P < .005) in those with vs those without subsequent pregnancy. Added to this body of data is the prospective POSITIVE trial, which showed that a temporary pause of endocrine therapy for an attempt at conceiving appears to be safe in young women with early HR+ breast cancer with short-term follow-up.4 Future research efforts investigating outcomes after assisted reproductive technologies in this population, those with germline mutations, and extended follow-up of studies, such as POSITIVE, will continue to inform guidance for and management of young women with breast cancer.

Guidelines favor the use of adjuvant chemotherapy for small, node-negative, triple-negative breast cancer (TNBC), specifically T1b and T1c tumors.5 However, high-quality data to inform this decision-making are sparse, and it is valuable to consider the magnitude of benefit weighed against possible risks and side effects of treatment, as well as patient comorbidities. A retrospective analysis of the Surveillance, Epidemiology, and End Results (SEER) database including 11,510 patients (3388 with T1b and 8122 with T1c TNBC) evaluated the impact of adjuvant chemotherapy on OS and breast cancer–specific survival (BCSS) (Carbajal-Ochoa et al). The use of adjuvant chemotherapy was associated with improved OS (hazard ratio 0.54; 95% CI 0.47-0.62; P < .001) and BCSS (hazard ratio 0.79; 95% CI 0.63-0.99; P = .043) among T1c TNBC. For those with T1b tumors, adjuvant chemotherapy improved OS (hazard ratio 0.52; 95% CI 0.41-0.68; P < .001) but did not improve BCSS (hazard ratio 0.70; 95% CI 0.45-1.07; P = .10). A better understanding of the molecular drivers implicated in this heterogeneous subtype, and predictors of response and resistance, will aid in identifying those patients who have greater benefit and those who can potentially be spared chemotherapy-related toxicities.

Additional References

  1. Anwar SL, Cahyono R, Prabowo D, et al. Metabolic comorbidities and the association with risks of recurrent metastatic disease in breast cancer survivors. BMC Cancer. 2021;21:590. doi: 10.1186/s12885-021-08343-0
  2. Sestak I, Distler W, Forbes JF, et al. Effect of body mass index on recurrences in tamoxifen and anastrozole treated women: An exploratory analysis from the ATAC trial. J Clin Oncol. 2010;28:3411-3415. doi: 10.1200/JCO.2009.27.2021
  3. Lambertini M, Blondeaux E, Bruzzone M, et al. Pregnancy after breast cancer: A systematic review and meta-analysis. J Clin Oncol. 2021;39:3293-3305. doi: 10.1200/JCO.21.00535
  4. Partridge AH, Niman SM, Ruggeri M, et al for the International Breast Cancer Study Group and POSITIVE Trial Collaborators. Interrupting endocrine therapy to attempt pregnancy after breast cancer. N Engl J Med. 2023;388:1645-1656. doi:10.1056/NEJMoa2212856
  5. Curigliano G, Burstein HJ, Winer EP, et al. De-escalating and escalating treatments for early-stage breast cancer: The St. Gallen International Expert Consensus Conference on the Primary Therapy of Early Breast Cancer 2017. Ann Oncol. 2017;28:1700-1712. doi:10.1093/annonc/mdx308
Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Publications
Topics
Sections
Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Dr. Roesch scans the journals so you don't have to!
Dr. Roesch scans the journals so you don't have to!

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Obesity and other metabolic comorbidities (including glucose intolerance, hypertension, and dyslipidemia) have been associated with poorer prognosis among breast cancer patients.1 The underlying mechanisms for which obesity is linked to inferior breast cancer outcomes is complex and may also involve drug efficacy in these patients. Data from the ATAC trial showed that there was a higher risk for recurrence among patients with obesity who were receiving an aromatase inhibitor (AI) vs patients with a healthy body weight receiving an AI; however, patients receiving tamoxifen did not exhibit this difference.2 A Danish Breast Cancer Group cohort study including 13,230 postmenopausal patients with stage I-III hormone receptor–positive (HR+) breast cancer treated with AI investigated the association of body mass index with recurrence (Harborg et al). There was a significantly increased risk for recurrence among those patients with obesity (adjusted hazard ratio 1.18; 95% CI 1.01-1.37) and severe obesity (adjusted hazard ratio 1.32; 95% CI 1.08-1.62) vs patients with healthy body weight. These results highlight the importance of lifestyle interventions targeting obesity and metabolic factors in breast cancer patients and support future studies investigating optimal drug selection based on body composition.

Breast cancer in young women presents a unique set of challenges owing to life-stage at the time of diagnosis and treatment. Oncofertility, family planning, and pregnancy are essential issues to address at the time of initial consultation and throughout the survivorship setting. Various studies have provided supportive evidence regarding the safety of pregnancy after breast cancer diagnosis and treatment.3 HR+ breast cancer is associated with its own distinctive considerations related to pregnancy and its timing, including the use of endocrine therapy for 5-10 years, the role of female hormones during pregnancy, and late patterns of recurrence that characterize this subtype. A meta-analysis including eight eligible studies and 3805 women with HR+ early breast cancer investigated the prognostic impact of future pregnancy among these patients (Arecco et al). A total of 1285 women had a pregnancy after breast cancer diagnosis and treatment; there was no difference in disease-free survival (hazard ratio 0.96; 95% CI 0.75-1.24; P = .781) and better overall survival (OS; hazard ratio 0.46; 95% CI 0.27-0.77; P < .005) in those with vs those without subsequent pregnancy. Added to this body of data is the prospective POSITIVE trial, which showed that a temporary pause of endocrine therapy for an attempt at conceiving appears to be safe in young women with early HR+ breast cancer with short-term follow-up.4 Future research efforts investigating outcomes after assisted reproductive technologies in this population, those with germline mutations, and extended follow-up of studies, such as POSITIVE, will continue to inform guidance for and management of young women with breast cancer.

Guidelines favor the use of adjuvant chemotherapy for small, node-negative, triple-negative breast cancer (TNBC), specifically T1b and T1c tumors.5 However, high-quality data to inform this decision-making are sparse, and it is valuable to consider the magnitude of benefit weighed against possible risks and side effects of treatment, as well as patient comorbidities. A retrospective analysis of the Surveillance, Epidemiology, and End Results (SEER) database including 11,510 patients (3388 with T1b and 8122 with T1c TNBC) evaluated the impact of adjuvant chemotherapy on OS and breast cancer–specific survival (BCSS) (Carbajal-Ochoa et al). The use of adjuvant chemotherapy was associated with improved OS (hazard ratio 0.54; 95% CI 0.47-0.62; P < .001) and BCSS (hazard ratio 0.79; 95% CI 0.63-0.99; P = .043) among T1c TNBC. For those with T1b tumors, adjuvant chemotherapy improved OS (hazard ratio 0.52; 95% CI 0.41-0.68; P < .001) but did not improve BCSS (hazard ratio 0.70; 95% CI 0.45-1.07; P = .10). A better understanding of the molecular drivers implicated in this heterogeneous subtype, and predictors of response and resistance, will aid in identifying those patients who have greater benefit and those who can potentially be spared chemotherapy-related toxicities.

Additional References

  1. Anwar SL, Cahyono R, Prabowo D, et al. Metabolic comorbidities and the association with risks of recurrent metastatic disease in breast cancer survivors. BMC Cancer. 2021;21:590. doi: 10.1186/s12885-021-08343-0
  2. Sestak I, Distler W, Forbes JF, et al. Effect of body mass index on recurrences in tamoxifen and anastrozole treated women: An exploratory analysis from the ATAC trial. J Clin Oncol. 2010;28:3411-3415. doi: 10.1200/JCO.2009.27.2021
  3. Lambertini M, Blondeaux E, Bruzzone M, et al. Pregnancy after breast cancer: A systematic review and meta-analysis. J Clin Oncol. 2021;39:3293-3305. doi: 10.1200/JCO.21.00535
  4. Partridge AH, Niman SM, Ruggeri M, et al for the International Breast Cancer Study Group and POSITIVE Trial Collaborators. Interrupting endocrine therapy to attempt pregnancy after breast cancer. N Engl J Med. 2023;388:1645-1656. doi:10.1056/NEJMoa2212856
  5. Curigliano G, Burstein HJ, Winer EP, et al. De-escalating and escalating treatments for early-stage breast cancer: The St. Gallen International Expert Consensus Conference on the Primary Therapy of Early Breast Cancer 2017. Ann Oncol. 2017;28:1700-1712. doi:10.1093/annonc/mdx308

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Obesity and other metabolic comorbidities (including glucose intolerance, hypertension, and dyslipidemia) have been associated with poorer prognosis among breast cancer patients.1 The underlying mechanisms for which obesity is linked to inferior breast cancer outcomes is complex and may also involve drug efficacy in these patients. Data from the ATAC trial showed that there was a higher risk for recurrence among patients with obesity who were receiving an aromatase inhibitor (AI) vs patients with a healthy body weight receiving an AI; however, patients receiving tamoxifen did not exhibit this difference.2 A Danish Breast Cancer Group cohort study including 13,230 postmenopausal patients with stage I-III hormone receptor–positive (HR+) breast cancer treated with AI investigated the association of body mass index with recurrence (Harborg et al). There was a significantly increased risk for recurrence among those patients with obesity (adjusted hazard ratio 1.18; 95% CI 1.01-1.37) and severe obesity (adjusted hazard ratio 1.32; 95% CI 1.08-1.62) vs patients with healthy body weight. These results highlight the importance of lifestyle interventions targeting obesity and metabolic factors in breast cancer patients and support future studies investigating optimal drug selection based on body composition.

Breast cancer in young women presents a unique set of challenges owing to life-stage at the time of diagnosis and treatment. Oncofertility, family planning, and pregnancy are essential issues to address at the time of initial consultation and throughout the survivorship setting. Various studies have provided supportive evidence regarding the safety of pregnancy after breast cancer diagnosis and treatment.3 HR+ breast cancer is associated with its own distinctive considerations related to pregnancy and its timing, including the use of endocrine therapy for 5-10 years, the role of female hormones during pregnancy, and late patterns of recurrence that characterize this subtype. A meta-analysis including eight eligible studies and 3805 women with HR+ early breast cancer investigated the prognostic impact of future pregnancy among these patients (Arecco et al). A total of 1285 women had a pregnancy after breast cancer diagnosis and treatment; there was no difference in disease-free survival (hazard ratio 0.96; 95% CI 0.75-1.24; P = .781) and better overall survival (OS; hazard ratio 0.46; 95% CI 0.27-0.77; P < .005) in those with vs those without subsequent pregnancy. Added to this body of data is the prospective POSITIVE trial, which showed that a temporary pause of endocrine therapy for an attempt at conceiving appears to be safe in young women with early HR+ breast cancer with short-term follow-up.4 Future research efforts investigating outcomes after assisted reproductive technologies in this population, those with germline mutations, and extended follow-up of studies, such as POSITIVE, will continue to inform guidance for and management of young women with breast cancer.

Guidelines favor the use of adjuvant chemotherapy for small, node-negative, triple-negative breast cancer (TNBC), specifically T1b and T1c tumors.5 However, high-quality data to inform this decision-making are sparse, and it is valuable to consider the magnitude of benefit weighed against possible risks and side effects of treatment, as well as patient comorbidities. A retrospective analysis of the Surveillance, Epidemiology, and End Results (SEER) database including 11,510 patients (3388 with T1b and 8122 with T1c TNBC) evaluated the impact of adjuvant chemotherapy on OS and breast cancer–specific survival (BCSS) (Carbajal-Ochoa et al). The use of adjuvant chemotherapy was associated with improved OS (hazard ratio 0.54; 95% CI 0.47-0.62; P < .001) and BCSS (hazard ratio 0.79; 95% CI 0.63-0.99; P = .043) among T1c TNBC. For those with T1b tumors, adjuvant chemotherapy improved OS (hazard ratio 0.52; 95% CI 0.41-0.68; P < .001) but did not improve BCSS (hazard ratio 0.70; 95% CI 0.45-1.07; P = .10). A better understanding of the molecular drivers implicated in this heterogeneous subtype, and predictors of response and resistance, will aid in identifying those patients who have greater benefit and those who can potentially be spared chemotherapy-related toxicities.

Additional References

  1. Anwar SL, Cahyono R, Prabowo D, et al. Metabolic comorbidities and the association with risks of recurrent metastatic disease in breast cancer survivors. BMC Cancer. 2021;21:590. doi: 10.1186/s12885-021-08343-0
  2. Sestak I, Distler W, Forbes JF, et al. Effect of body mass index on recurrences in tamoxifen and anastrozole treated women: An exploratory analysis from the ATAC trial. J Clin Oncol. 2010;28:3411-3415. doi: 10.1200/JCO.2009.27.2021
  3. Lambertini M, Blondeaux E, Bruzzone M, et al. Pregnancy after breast cancer: A systematic review and meta-analysis. J Clin Oncol. 2021;39:3293-3305. doi: 10.1200/JCO.21.00535
  4. Partridge AH, Niman SM, Ruggeri M, et al for the International Breast Cancer Study Group and POSITIVE Trial Collaborators. Interrupting endocrine therapy to attempt pregnancy after breast cancer. N Engl J Med. 2023;388:1645-1656. doi:10.1056/NEJMoa2212856
  5. Curigliano G, Burstein HJ, Winer EP, et al. De-escalating and escalating treatments for early-stage breast cancer: The St. Gallen International Expert Consensus Conference on the Primary Therapy of Early Breast Cancer 2017. Ann Oncol. 2017;28:1700-1712. doi:10.1093/annonc/mdx308
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Fri, 09/29/2023 - 14:00
Un-Gate On Date
Fri, 09/29/2023 - 14:00
Use ProPublica
CFC Schedule Remove Status
Fri, 09/29/2023 - 14:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Commentary: Axillary Surgery, PM2.5, and Treatment With Tucatinib in Breast Cancer, November 2023

Article Type
Changed
Thu, 11/09/2023 - 11:58
Display Headline
Commentary: Axillary Surgery, PM2.5, and Treatment With Tucatinib in Breast Cancer, November 2023
Dr. Roesch scans the journals so you don't have to!

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Support for axillary surgery de-escalation for select patients with early-stage breast cancer has been demonstrated in prior studies,1,2 leading to widespread use of sentinel lymph node biopsy (SLNB) for axillary staging for many patients. For example, the phase 3 randomized ACOSOG Z0011 trial showed that among women with T1/2 breast cancer, without palpable lymph nodes and one to two sentinel lymph nodes positive, survival outcomes were noninferior for sentinel lymph node dissection vs axillary lymph node dissection.1 The SOUND (Sentinel Node vs Observation After Axillary Ultra-Sound) trial was a phase 3 prospective randomized study that included 1405 women with early breast cancer, tumor size ≤ 2 cm, and negative preoperative axillary ultrasound, and was designed to investigate the effect of axillary surgery omission in these patients (Gentilini et al). Five-year distant disease-free survival, the primary endpoint, was 97.7% in the SLNB group and 98.0% in the no-axillary-surgery group (log-rank P = .67; hazard ratio [HR] 0.84, noninferiority P = .02). Rates of locoregional relapse (1.7% vs 1.6%), distant metastases (1.8% vs 2.0%), and deaths (3.0% vs 2.6%) were similar in the SLNB group compared with the no-axillary-surgery group, respectively. Furthermore, adjuvant treatments were not significantly different between the two groups, indicating that tumor biology/genomics may have an expanding role in tailoring adjuvant therapy compared with clinicopathologic features. The results of this study suggest that axillary surgery omission can be considered in patients with ≤ T2 early breast cancer and negative axillary ultrasound when absence of this pathologic information does not affect the adjuvant treatment plan.

Hormone receptor–positive breast cancer is the most common subtype, with established risk factors including exposure to exogenous hormones, reproductive history, and lifestyle components (alcohol intake, obesity). There are also less-recognized environmental influences that may disrupt endocrine pathways and, as a result, affect tumor development. Fine particulate matter (PM2.5), produced by combustion processes (vehicles, industrial facilities), burning wood, and fires, among other sources, is composed of various airborne pollutants (metals, organic compounds, ammonium, nitrate, ozone, sulfate, etc.). Prior studies evaluating the association of PM2.5 and breast cancer development have shown mixed results.3,4 A prospective US cohort study including 196,905 women without a prior history of breast cancer estimated historical annual average PM2.5 concentrations between 1980 and 1984 (10 years prior to enrollment) (White et al). A total of 15,870 breast cancer cases were identified, and a 10 μg/m3 increase in PM2.5 was associated with an 8% increase in overall breast cancer incidence (HR 1.08; 95% CI 1.02-1.13). The association was observed for estrogen-receptor (ER)-positive (HR 1.10; 95% CI 1.04-1.17) but not ER-negative tumors. Future studies focusing on historic exposures, investigating geographic differences and the resultant effect on cancer development, are of interest.

HER2CLIMB was a pivotal phase 3 randomized, double-blinded trial that demonstrated significant improvement in survival outcomes with the combination of tucatinib/trastuzumab/capecitabine vs tucatinib/trastuzumab/placebo among patients with previously treated human epidermal growth factor receptor 2 (HER2)-positive metastatic breast cancer.5 Real-world data help inform our daily practice because patients enrolled in clinical trials do not always accurately represent the general population. A retrospective cohort study including 3449 patients with HER2-positive metastatic breast cancer evaluated outcomes with tucatinib in a real-world setting, demonstrating results similar to those seen in HER2CLIMB. Among all patients who received tucatinib (n = 216), median real-world time to treatment discontinuation was 6.5 months (95% CI 5.4-8.8), median real-world time to next treatment (which can serve as a proxy for progression-free survival) was 8.7 months (95% CI 6.8-10.7), and real-world overall survival was 26.6 months (95% CI 20.2–not reached). Median real-world time to treatment discontinuation was 8.1 months (95% CI 5.7-9.5) for patients who received the approved tucatinib triplet combination after one or more HER2-directed regimens in the metastatic setting and 9.4 months (95% CI 6.3-14.1) for those receiving it in the second- or third-line setting (Kaufman et al). These results support the efficacy of tucatinib in a real-world population, suggesting that earlier use (second or third line) may result in better outcomes. Future studies will continue to address the positioning of tucatinib in the treatment algorithm for HER2-positive metastatic breast cancer, including the evaluation of novel combinations.

Additional References

  1. Giuliano AE et al. Effect of axillary dissection vs no axillary dissection on 10-year overall survival among women with invasive breast cancer and sentinel node metastasis: The ACOSOG Z0011 (Alliance) randomized clinical trial. JAMA. 2017;318:918-926. doi: 10.1001/jama.2017.11470 
  2. Bartels SAL, Donker M, et al. Radiotherapy or surgery of the axilla after a positive sentinel node in breast cancer: 10-year results of the randomized controlled EORTC 10981-22023 AMAROS Trial. J Clin Oncol. 2023;41:2159-2165. doi: 10.1200/JCO.22.01565
  3. Gabet S, Lemarchand C, Guénel P, Slama R. Breast cancer risk in association with atmospheric pollution exposure: A meta-analysis of effect estimates followed by a health impact assessment. Environ Health Perspect. 2021;129:57012. doi: 10.1289/EHP8419
  4. Hvidtfeldt UA et al. Breast cancer incidence in relation to long-term low-level exposure to air pollution in the ELAPSE pooled cohort. Cancer Epidemiol Biomarkers Prev. 2023;32:105-113. doi: 10.1158/1055-9965.EPI-22-0720  
  5. Murthy RK et al. Tucatinib, trastuzumab, and capecitabine for HER2-positive metastatic breast cancer. N Engl J Med. 2020;382:597-609. doi:10.1056/NEJMoa1914609
Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Publications
Topics
Sections
Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Dr. Roesch scans the journals so you don't have to!
Dr. Roesch scans the journals so you don't have to!

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Support for axillary surgery de-escalation for select patients with early-stage breast cancer has been demonstrated in prior studies,1,2 leading to widespread use of sentinel lymph node biopsy (SLNB) for axillary staging for many patients. For example, the phase 3 randomized ACOSOG Z0011 trial showed that among women with T1/2 breast cancer, without palpable lymph nodes and one to two sentinel lymph nodes positive, survival outcomes were noninferior for sentinel lymph node dissection vs axillary lymph node dissection.1 The SOUND (Sentinel Node vs Observation After Axillary Ultra-Sound) trial was a phase 3 prospective randomized study that included 1405 women with early breast cancer, tumor size ≤ 2 cm, and negative preoperative axillary ultrasound, and was designed to investigate the effect of axillary surgery omission in these patients (Gentilini et al). Five-year distant disease-free survival, the primary endpoint, was 97.7% in the SLNB group and 98.0% in the no-axillary-surgery group (log-rank P = .67; hazard ratio [HR] 0.84, noninferiority P = .02). Rates of locoregional relapse (1.7% vs 1.6%), distant metastases (1.8% vs 2.0%), and deaths (3.0% vs 2.6%) were similar in the SLNB group compared with the no-axillary-surgery group, respectively. Furthermore, adjuvant treatments were not significantly different between the two groups, indicating that tumor biology/genomics may have an expanding role in tailoring adjuvant therapy compared with clinicopathologic features. The results of this study suggest that axillary surgery omission can be considered in patients with ≤ T2 early breast cancer and negative axillary ultrasound when absence of this pathologic information does not affect the adjuvant treatment plan.

Hormone receptor–positive breast cancer is the most common subtype, with established risk factors including exposure to exogenous hormones, reproductive history, and lifestyle components (alcohol intake, obesity). There are also less-recognized environmental influences that may disrupt endocrine pathways and, as a result, affect tumor development. Fine particulate matter (PM2.5), produced by combustion processes (vehicles, industrial facilities), burning wood, and fires, among other sources, is composed of various airborne pollutants (metals, organic compounds, ammonium, nitrate, ozone, sulfate, etc.). Prior studies evaluating the association of PM2.5 and breast cancer development have shown mixed results.3,4 A prospective US cohort study including 196,905 women without a prior history of breast cancer estimated historical annual average PM2.5 concentrations between 1980 and 1984 (10 years prior to enrollment) (White et al). A total of 15,870 breast cancer cases were identified, and a 10 μg/m3 increase in PM2.5 was associated with an 8% increase in overall breast cancer incidence (HR 1.08; 95% CI 1.02-1.13). The association was observed for estrogen-receptor (ER)-positive (HR 1.10; 95% CI 1.04-1.17) but not ER-negative tumors. Future studies focusing on historic exposures, investigating geographic differences and the resultant effect on cancer development, are of interest.

HER2CLIMB was a pivotal phase 3 randomized, double-blinded trial that demonstrated significant improvement in survival outcomes with the combination of tucatinib/trastuzumab/capecitabine vs tucatinib/trastuzumab/placebo among patients with previously treated human epidermal growth factor receptor 2 (HER2)-positive metastatic breast cancer.5 Real-world data help inform our daily practice because patients enrolled in clinical trials do not always accurately represent the general population. A retrospective cohort study including 3449 patients with HER2-positive metastatic breast cancer evaluated outcomes with tucatinib in a real-world setting, demonstrating results similar to those seen in HER2CLIMB. Among all patients who received tucatinib (n = 216), median real-world time to treatment discontinuation was 6.5 months (95% CI 5.4-8.8), median real-world time to next treatment (which can serve as a proxy for progression-free survival) was 8.7 months (95% CI 6.8-10.7), and real-world overall survival was 26.6 months (95% CI 20.2–not reached). Median real-world time to treatment discontinuation was 8.1 months (95% CI 5.7-9.5) for patients who received the approved tucatinib triplet combination after one or more HER2-directed regimens in the metastatic setting and 9.4 months (95% CI 6.3-14.1) for those receiving it in the second- or third-line setting (Kaufman et al). These results support the efficacy of tucatinib in a real-world population, suggesting that earlier use (second or third line) may result in better outcomes. Future studies will continue to address the positioning of tucatinib in the treatment algorithm for HER2-positive metastatic breast cancer, including the evaluation of novel combinations.

Additional References

  1. Giuliano AE et al. Effect of axillary dissection vs no axillary dissection on 10-year overall survival among women with invasive breast cancer and sentinel node metastasis: The ACOSOG Z0011 (Alliance) randomized clinical trial. JAMA. 2017;318:918-926. doi: 10.1001/jama.2017.11470 
  2. Bartels SAL, Donker M, et al. Radiotherapy or surgery of the axilla after a positive sentinel node in breast cancer: 10-year results of the randomized controlled EORTC 10981-22023 AMAROS Trial. J Clin Oncol. 2023;41:2159-2165. doi: 10.1200/JCO.22.01565
  3. Gabet S, Lemarchand C, Guénel P, Slama R. Breast cancer risk in association with atmospheric pollution exposure: A meta-analysis of effect estimates followed by a health impact assessment. Environ Health Perspect. 2021;129:57012. doi: 10.1289/EHP8419
  4. Hvidtfeldt UA et al. Breast cancer incidence in relation to long-term low-level exposure to air pollution in the ELAPSE pooled cohort. Cancer Epidemiol Biomarkers Prev. 2023;32:105-113. doi: 10.1158/1055-9965.EPI-22-0720  
  5. Murthy RK et al. Tucatinib, trastuzumab, and capecitabine for HER2-positive metastatic breast cancer. N Engl J Med. 2020;382:597-609. doi:10.1056/NEJMoa1914609

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Support for axillary surgery de-escalation for select patients with early-stage breast cancer has been demonstrated in prior studies,1,2 leading to widespread use of sentinel lymph node biopsy (SLNB) for axillary staging for many patients. For example, the phase 3 randomized ACOSOG Z0011 trial showed that among women with T1/2 breast cancer, without palpable lymph nodes and one to two sentinel lymph nodes positive, survival outcomes were noninferior for sentinel lymph node dissection vs axillary lymph node dissection.1 The SOUND (Sentinel Node vs Observation After Axillary Ultra-Sound) trial was a phase 3 prospective randomized study that included 1405 women with early breast cancer, tumor size ≤ 2 cm, and negative preoperative axillary ultrasound, and was designed to investigate the effect of axillary surgery omission in these patients (Gentilini et al). Five-year distant disease-free survival, the primary endpoint, was 97.7% in the SLNB group and 98.0% in the no-axillary-surgery group (log-rank P = .67; hazard ratio [HR] 0.84, noninferiority P = .02). Rates of locoregional relapse (1.7% vs 1.6%), distant metastases (1.8% vs 2.0%), and deaths (3.0% vs 2.6%) were similar in the SLNB group compared with the no-axillary-surgery group, respectively. Furthermore, adjuvant treatments were not significantly different between the two groups, indicating that tumor biology/genomics may have an expanding role in tailoring adjuvant therapy compared with clinicopathologic features. The results of this study suggest that axillary surgery omission can be considered in patients with ≤ T2 early breast cancer and negative axillary ultrasound when absence of this pathologic information does not affect the adjuvant treatment plan.

Hormone receptor–positive breast cancer is the most common subtype, with established risk factors including exposure to exogenous hormones, reproductive history, and lifestyle components (alcohol intake, obesity). There are also less-recognized environmental influences that may disrupt endocrine pathways and, as a result, affect tumor development. Fine particulate matter (PM2.5), produced by combustion processes (vehicles, industrial facilities), burning wood, and fires, among other sources, is composed of various airborne pollutants (metals, organic compounds, ammonium, nitrate, ozone, sulfate, etc.). Prior studies evaluating the association of PM2.5 and breast cancer development have shown mixed results.3,4 A prospective US cohort study including 196,905 women without a prior history of breast cancer estimated historical annual average PM2.5 concentrations between 1980 and 1984 (10 years prior to enrollment) (White et al). A total of 15,870 breast cancer cases were identified, and a 10 μg/m3 increase in PM2.5 was associated with an 8% increase in overall breast cancer incidence (HR 1.08; 95% CI 1.02-1.13). The association was observed for estrogen-receptor (ER)-positive (HR 1.10; 95% CI 1.04-1.17) but not ER-negative tumors. Future studies focusing on historic exposures, investigating geographic differences and the resultant effect on cancer development, are of interest.

HER2CLIMB was a pivotal phase 3 randomized, double-blinded trial that demonstrated significant improvement in survival outcomes with the combination of tucatinib/trastuzumab/capecitabine vs tucatinib/trastuzumab/placebo among patients with previously treated human epidermal growth factor receptor 2 (HER2)-positive metastatic breast cancer.5 Real-world data help inform our daily practice because patients enrolled in clinical trials do not always accurately represent the general population. A retrospective cohort study including 3449 patients with HER2-positive metastatic breast cancer evaluated outcomes with tucatinib in a real-world setting, demonstrating results similar to those seen in HER2CLIMB. Among all patients who received tucatinib (n = 216), median real-world time to treatment discontinuation was 6.5 months (95% CI 5.4-8.8), median real-world time to next treatment (which can serve as a proxy for progression-free survival) was 8.7 months (95% CI 6.8-10.7), and real-world overall survival was 26.6 months (95% CI 20.2–not reached). Median real-world time to treatment discontinuation was 8.1 months (95% CI 5.7-9.5) for patients who received the approved tucatinib triplet combination after one or more HER2-directed regimens in the metastatic setting and 9.4 months (95% CI 6.3-14.1) for those receiving it in the second- or third-line setting (Kaufman et al). These results support the efficacy of tucatinib in a real-world population, suggesting that earlier use (second or third line) may result in better outcomes. Future studies will continue to address the positioning of tucatinib in the treatment algorithm for HER2-positive metastatic breast cancer, including the evaluation of novel combinations.

Additional References

  1. Giuliano AE et al. Effect of axillary dissection vs no axillary dissection on 10-year overall survival among women with invasive breast cancer and sentinel node metastasis: The ACOSOG Z0011 (Alliance) randomized clinical trial. JAMA. 2017;318:918-926. doi: 10.1001/jama.2017.11470 
  2. Bartels SAL, Donker M, et al. Radiotherapy or surgery of the axilla after a positive sentinel node in breast cancer: 10-year results of the randomized controlled EORTC 10981-22023 AMAROS Trial. J Clin Oncol. 2023;41:2159-2165. doi: 10.1200/JCO.22.01565
  3. Gabet S, Lemarchand C, Guénel P, Slama R. Breast cancer risk in association with atmospheric pollution exposure: A meta-analysis of effect estimates followed by a health impact assessment. Environ Health Perspect. 2021;129:57012. doi: 10.1289/EHP8419
  4. Hvidtfeldt UA et al. Breast cancer incidence in relation to long-term low-level exposure to air pollution in the ELAPSE pooled cohort. Cancer Epidemiol Biomarkers Prev. 2023;32:105-113. doi: 10.1158/1055-9965.EPI-22-0720  
  5. Murthy RK et al. Tucatinib, trastuzumab, and capecitabine for HER2-positive metastatic breast cancer. N Engl J Med. 2020;382:597-609. doi:10.1056/NEJMoa1914609
Publications
Publications
Topics
Article Type
Display Headline
Commentary: Axillary Surgery, PM2.5, and Treatment With Tucatinib in Breast Cancer, November 2023
Display Headline
Commentary: Axillary Surgery, PM2.5, and Treatment With Tucatinib in Breast Cancer, November 2023
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Fri, 09/29/2023 - 14:00
Un-Gate On Date
Fri, 09/29/2023 - 14:00
Use ProPublica
CFC Schedule Remove Status
Fri, 09/29/2023 - 14:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Activity Salesforce Deliverable ID
367005.2
Activity ID
93656
Product Name
Clinical Edge Journal Scan
Product ID
124
Supporter Name /ID
Perjeta [ 3532 ]

Commentary: Radiation therapy, endocrine therapy, metformin, and statins in breast cancer, October 2023

Article Type
Changed
Wed, 11/01/2023 - 21:59
Dr. Roesch scans the journals so you don't have to!

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A11.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Radiation therapy (RT) is typically administered after breast-conserving surgery (BCS) and has demonstrated reduction in local recurrence risk. A recent phase 3 trial evaluating the omission of radiation among patients aged 65 years or older with hormone receptor-positive, node-negative, T1/2 tumors (≤ 3 cm) treated with BCS and adjuvant endocrine therapy, showed an increased incidence of local recurrence in the no-RT group compared with the RT group (9.5% vs 0.9% within 10 years) but no difference in distant recurrence or overall survival.1 With advancements in the field of precision medicine, it has been postulated that combining molecular biomarkers and clinicopathologic features can more distinctly identify patients with low-risk disease for whom radiation can be omitted. A prospective cohort study including 500 women aged 55 years or older with T1N0, grade 1 or 2, luminal A–subtype breast cancer (estrogen-receptor positivity [ER+]  > 1%, progesterone receptor positivity > 20%, human epidermal growth factor receptor 2–negative [HER2-], Ki67 index ≤ 13.25%) after BCS and adjuvant endocrine therapy demonstrated low rates of local recurrence with the omission of RT (Whelan et al). At 5 years, the cumulative incidence of local recurrence was 2.3% (90% CI 1.3-3.8; 95% CI 1.2-4.1), contralateral breast recurrence was 1.9% (90% CI 1.1-3.2), and recurrence of any type was 2.7% (90% CI 1.6-4.1). This study supports consideration of a local therapy de-escalation approach, specifically RT omission, for select patients with tumors characterized by favorable molecular and clinical features.

Endocrine therapy (ET) remains the backbone of treatment for hormone receptor–positive breast cancer; however, 15%-20% of tumors are initially resistant to ET and endocrine resistance develops over time in approximately 30%-40%.2 In an effort to overcome limitations with historical standard-of-care endocrine agents, the class of oral potent selective estrogen receptor degraders (SERD) is evolving. The phase 2, randomized, controlled coopERA Breast Cancer trial evaluated the antiproliferative effect of giredestrant (a highly potent nonsteroidal oral SERD) compared with anastrozole (each combined with palbociclib after 2-week window-of-opportunity phase) among postmenopausal women with early-stage (cT1c-cT4) ER+/HER2- breast cancer with a Ki67 score ≥ 5% (Hurvitz et al). Among 221 enrolled patients (giredestrant group n = 112, and anastrozole group n = 109), giredestrant led to a significantly greater relative geometric mean reduction of Ki67 at 2 weeks from baseline compared with anastrozole (-75% vs -67%; P = 0.043). Neutropenia (26% and 27%) and decreased neutrophil count (15% and 15%) were the most common grade 3-4 adverse events in the giredestrant-palbociclib and anastrozole-palbociclib groups, respectively. The value of Ki67 as a biomarker for efficacy and outcome was demonstrated in the phase 3 POETIC trial, which showed that the degree of Ki67 reduction after 2 weeks of ET correlated with 5-year recurrence risk.3 These data encourage further investigation of oral SERD combinations, predictors of response, and long-term outcomes that may influence agent selection and sequencing.

Anticancer properties have been demonstrated with aspirin, statins, and metformin, although the data on the prognostic impact of these agents in breast cancer have shown mixed results.4 A nationwide population-based cohort study including 26,190 women aged 50 years or older diagnosed with breast cancer and surviving 12 months or more after diagnosis was performed to evaluate the postdiagnosis use of aspirin, statins, and metformin and association with breast cancer-specific survival (BCSS) (Löfling et al). At 6.1 years of follow-up, there were 2169 deaths related to breast cancer and the results supported an association of postdiagnostic use of statins and metformin with survival (hazard ratio for association between use of statins vs no use and BCSS was 0.84 [95% CI 0.75-0.94]; hazard ratio for association between metformin use vs use of nonmetformin antidiabetics and BCSS was 0.70 [95% CI 0.51-0.96]). Furthermore, there appeared to be differences in association by ER status. An important relationship exists between cardiovascular health and breast cancer, and future efforts should continue to study pharmacologic and lifestyle interventions that may optimize metabolic profiles and improve outcomes for patients.

Additional References

  1. Kunkler IH, Williams LJ, Jack WJL, et al. Breast-conserving surgery with or without irradiation in early breast cancer. N Engl J Med. 2023;388:585-594. doi: 10.1056/NEJMoa2207586  
  2. Lei JT, Anurag M, Haricharan S, et al. Endocrine therapy resistance: New insights. Breast. 2019;48:S26-S30. doi: 10.1016/S0960-9776(19)31118-X  
  3. Smith I, Robertson J, Kilburn L, et al. Long-term outcome and prognostic value of Ki67 after perioperative endocrine therapy in postmenopausal women with hormone-sensitive early breast cancer (POETIC): An open-label, multicentre, parallel-group, randomised, phase 3 trial. Lancet Oncol. 2020;21:1443-1454. doi: 10.1016/S1470-2045(20)30458-7
  4. Nowakowska MK, Lei X, Thompson MT, et al. Association of statin use with clinical outcomes in patients with triple-negative breast cancer. Cancer. 2021;127:4142-4150. doi: 10.1002/cncr.33797
Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Publications
Topics
Sections
Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Dr. Roesch scans the journals so you don't have to!
Dr. Roesch scans the journals so you don't have to!

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A11.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Radiation therapy (RT) is typically administered after breast-conserving surgery (BCS) and has demonstrated reduction in local recurrence risk. A recent phase 3 trial evaluating the omission of radiation among patients aged 65 years or older with hormone receptor-positive, node-negative, T1/2 tumors (≤ 3 cm) treated with BCS and adjuvant endocrine therapy, showed an increased incidence of local recurrence in the no-RT group compared with the RT group (9.5% vs 0.9% within 10 years) but no difference in distant recurrence or overall survival.1 With advancements in the field of precision medicine, it has been postulated that combining molecular biomarkers and clinicopathologic features can more distinctly identify patients with low-risk disease for whom radiation can be omitted. A prospective cohort study including 500 women aged 55 years or older with T1N0, grade 1 or 2, luminal A–subtype breast cancer (estrogen-receptor positivity [ER+]  > 1%, progesterone receptor positivity > 20%, human epidermal growth factor receptor 2–negative [HER2-], Ki67 index ≤ 13.25%) after BCS and adjuvant endocrine therapy demonstrated low rates of local recurrence with the omission of RT (Whelan et al). At 5 years, the cumulative incidence of local recurrence was 2.3% (90% CI 1.3-3.8; 95% CI 1.2-4.1), contralateral breast recurrence was 1.9% (90% CI 1.1-3.2), and recurrence of any type was 2.7% (90% CI 1.6-4.1). This study supports consideration of a local therapy de-escalation approach, specifically RT omission, for select patients with tumors characterized by favorable molecular and clinical features.

Endocrine therapy (ET) remains the backbone of treatment for hormone receptor–positive breast cancer; however, 15%-20% of tumors are initially resistant to ET and endocrine resistance develops over time in approximately 30%-40%.2 In an effort to overcome limitations with historical standard-of-care endocrine agents, the class of oral potent selective estrogen receptor degraders (SERD) is evolving. The phase 2, randomized, controlled coopERA Breast Cancer trial evaluated the antiproliferative effect of giredestrant (a highly potent nonsteroidal oral SERD) compared with anastrozole (each combined with palbociclib after 2-week window-of-opportunity phase) among postmenopausal women with early-stage (cT1c-cT4) ER+/HER2- breast cancer with a Ki67 score ≥ 5% (Hurvitz et al). Among 221 enrolled patients (giredestrant group n = 112, and anastrozole group n = 109), giredestrant led to a significantly greater relative geometric mean reduction of Ki67 at 2 weeks from baseline compared with anastrozole (-75% vs -67%; P = 0.043). Neutropenia (26% and 27%) and decreased neutrophil count (15% and 15%) were the most common grade 3-4 adverse events in the giredestrant-palbociclib and anastrozole-palbociclib groups, respectively. The value of Ki67 as a biomarker for efficacy and outcome was demonstrated in the phase 3 POETIC trial, which showed that the degree of Ki67 reduction after 2 weeks of ET correlated with 5-year recurrence risk.3 These data encourage further investigation of oral SERD combinations, predictors of response, and long-term outcomes that may influence agent selection and sequencing.

Anticancer properties have been demonstrated with aspirin, statins, and metformin, although the data on the prognostic impact of these agents in breast cancer have shown mixed results.4 A nationwide population-based cohort study including 26,190 women aged 50 years or older diagnosed with breast cancer and surviving 12 months or more after diagnosis was performed to evaluate the postdiagnosis use of aspirin, statins, and metformin and association with breast cancer-specific survival (BCSS) (Löfling et al). At 6.1 years of follow-up, there were 2169 deaths related to breast cancer and the results supported an association of postdiagnostic use of statins and metformin with survival (hazard ratio for association between use of statins vs no use and BCSS was 0.84 [95% CI 0.75-0.94]; hazard ratio for association between metformin use vs use of nonmetformin antidiabetics and BCSS was 0.70 [95% CI 0.51-0.96]). Furthermore, there appeared to be differences in association by ER status. An important relationship exists between cardiovascular health and breast cancer, and future efforts should continue to study pharmacologic and lifestyle interventions that may optimize metabolic profiles and improve outcomes for patients.

Additional References

  1. Kunkler IH, Williams LJ, Jack WJL, et al. Breast-conserving surgery with or without irradiation in early breast cancer. N Engl J Med. 2023;388:585-594. doi: 10.1056/NEJMoa2207586  
  2. Lei JT, Anurag M, Haricharan S, et al. Endocrine therapy resistance: New insights. Breast. 2019;48:S26-S30. doi: 10.1016/S0960-9776(19)31118-X  
  3. Smith I, Robertson J, Kilburn L, et al. Long-term outcome and prognostic value of Ki67 after perioperative endocrine therapy in postmenopausal women with hormone-sensitive early breast cancer (POETIC): An open-label, multicentre, parallel-group, randomised, phase 3 trial. Lancet Oncol. 2020;21:1443-1454. doi: 10.1016/S1470-2045(20)30458-7
  4. Nowakowska MK, Lei X, Thompson MT, et al. Association of statin use with clinical outcomes in patients with triple-negative breast cancer. Cancer. 2021;127:4142-4150. doi: 10.1002/cncr.33797

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A11.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Radiation therapy (RT) is typically administered after breast-conserving surgery (BCS) and has demonstrated reduction in local recurrence risk. A recent phase 3 trial evaluating the omission of radiation among patients aged 65 years or older with hormone receptor-positive, node-negative, T1/2 tumors (≤ 3 cm) treated with BCS and adjuvant endocrine therapy, showed an increased incidence of local recurrence in the no-RT group compared with the RT group (9.5% vs 0.9% within 10 years) but no difference in distant recurrence or overall survival.1 With advancements in the field of precision medicine, it has been postulated that combining molecular biomarkers and clinicopathologic features can more distinctly identify patients with low-risk disease for whom radiation can be omitted. A prospective cohort study including 500 women aged 55 years or older with T1N0, grade 1 or 2, luminal A–subtype breast cancer (estrogen-receptor positivity [ER+]  > 1%, progesterone receptor positivity > 20%, human epidermal growth factor receptor 2–negative [HER2-], Ki67 index ≤ 13.25%) after BCS and adjuvant endocrine therapy demonstrated low rates of local recurrence with the omission of RT (Whelan et al). At 5 years, the cumulative incidence of local recurrence was 2.3% (90% CI 1.3-3.8; 95% CI 1.2-4.1), contralateral breast recurrence was 1.9% (90% CI 1.1-3.2), and recurrence of any type was 2.7% (90% CI 1.6-4.1). This study supports consideration of a local therapy de-escalation approach, specifically RT omission, for select patients with tumors characterized by favorable molecular and clinical features.

Endocrine therapy (ET) remains the backbone of treatment for hormone receptor–positive breast cancer; however, 15%-20% of tumors are initially resistant to ET and endocrine resistance develops over time in approximately 30%-40%.2 In an effort to overcome limitations with historical standard-of-care endocrine agents, the class of oral potent selective estrogen receptor degraders (SERD) is evolving. The phase 2, randomized, controlled coopERA Breast Cancer trial evaluated the antiproliferative effect of giredestrant (a highly potent nonsteroidal oral SERD) compared with anastrozole (each combined with palbociclib after 2-week window-of-opportunity phase) among postmenopausal women with early-stage (cT1c-cT4) ER+/HER2- breast cancer with a Ki67 score ≥ 5% (Hurvitz et al). Among 221 enrolled patients (giredestrant group n = 112, and anastrozole group n = 109), giredestrant led to a significantly greater relative geometric mean reduction of Ki67 at 2 weeks from baseline compared with anastrozole (-75% vs -67%; P = 0.043). Neutropenia (26% and 27%) and decreased neutrophil count (15% and 15%) were the most common grade 3-4 adverse events in the giredestrant-palbociclib and anastrozole-palbociclib groups, respectively. The value of Ki67 as a biomarker for efficacy and outcome was demonstrated in the phase 3 POETIC trial, which showed that the degree of Ki67 reduction after 2 weeks of ET correlated with 5-year recurrence risk.3 These data encourage further investigation of oral SERD combinations, predictors of response, and long-term outcomes that may influence agent selection and sequencing.

Anticancer properties have been demonstrated with aspirin, statins, and metformin, although the data on the prognostic impact of these agents in breast cancer have shown mixed results.4 A nationwide population-based cohort study including 26,190 women aged 50 years or older diagnosed with breast cancer and surviving 12 months or more after diagnosis was performed to evaluate the postdiagnosis use of aspirin, statins, and metformin and association with breast cancer-specific survival (BCSS) (Löfling et al). At 6.1 years of follow-up, there were 2169 deaths related to breast cancer and the results supported an association of postdiagnostic use of statins and metformin with survival (hazard ratio for association between use of statins vs no use and BCSS was 0.84 [95% CI 0.75-0.94]; hazard ratio for association between metformin use vs use of nonmetformin antidiabetics and BCSS was 0.70 [95% CI 0.51-0.96]). Furthermore, there appeared to be differences in association by ER status. An important relationship exists between cardiovascular health and breast cancer, and future efforts should continue to study pharmacologic and lifestyle interventions that may optimize metabolic profiles and improve outcomes for patients.

Additional References

  1. Kunkler IH, Williams LJ, Jack WJL, et al. Breast-conserving surgery with or without irradiation in early breast cancer. N Engl J Med. 2023;388:585-594. doi: 10.1056/NEJMoa2207586  
  2. Lei JT, Anurag M, Haricharan S, et al. Endocrine therapy resistance: New insights. Breast. 2019;48:S26-S30. doi: 10.1016/S0960-9776(19)31118-X  
  3. Smith I, Robertson J, Kilburn L, et al. Long-term outcome and prognostic value of Ki67 after perioperative endocrine therapy in postmenopausal women with hormone-sensitive early breast cancer (POETIC): An open-label, multicentre, parallel-group, randomised, phase 3 trial. Lancet Oncol. 2020;21:1443-1454. doi: 10.1016/S1470-2045(20)30458-7
  4. Nowakowska MK, Lei X, Thompson MT, et al. Association of statin use with clinical outcomes in patients with triple-negative breast cancer. Cancer. 2021;127:4142-4150. doi: 10.1002/cncr.33797
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Fri, 09/29/2023 - 14:00
Un-Gate On Date
Fri, 09/29/2023 - 14:00
Use ProPublica
CFC Schedule Remove Status
Fri, 09/29/2023 - 14:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Activity Salesforce Deliverable ID
367005.2
Activity ID
93656
Product Name
Clinical Edge Journal Scan
Product ID
124
Supporter Name /ID
Perjeta [ 3532 ]

Commentary: Age and breast cancer, and cardiometabolic comorbidities, September 2023

Article Type
Changed
Tue, 09/19/2023 - 12:49
Dr. Roesch scans the journals, so you don't have to!

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A11.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Overdiagnosis — defined as cancer detection, often through screening, that would not have led to symptoms during one’s lifetime — can be an issue associated with breast cancer screening in older women. Observational data have shown that continuing screening past age 75 years does not lead to substantial reductions in breast cancer mortality.1 A retrospective cohort study using the Surveillance, Epidemiology, and End Results (SEER)-Medicare registry, including 54,635 women ≥ 70 years of age, compared the cumulative incidence of breast cancer among older women who continued screening with those who did not and demonstrated that the relative risk for overdiagnosis increases with older age and lower life expectancy (Richman et al). The cumulative incidence of breast cancer was 6.1 vs 4.2 cases per 100 screened vs unscreened women among those aged 70-74 years, with an estimated 31% potentially overdiagnosed in the screened group. For women aged 75-84 years and ≥ 85 years, the estimated rates of breast cancer overdiagnosis were 47% and 54%, respectively. Furthermore, screening did not lead to a statistically significant difference in breast cancer–specific mortality in any of these age groups. The risks and benefits of breast cancer screening should be fully discussed with patients, and this decision-making process should consider a woman’s preference, comorbidities, and willingness to undergo specific treatments.

Studies have shown that breast cancer survivors have increased rates of age-related conditions, including cardiovascular disease and osteoporosis among others, therefore postulating that the biological aging process may be accelerated in this population.2 Among 417 women enrolled in the prospective Sister Study cohort, paired blood samples collected an average of 7.7 years apart compared three epigenetic metrics of biological aging (calculated on the basis of DNA methylation data) between women who were diagnosed and treated for breast cancer (n = 190) vs those who remained breast cancer–free (n = 227) (Kresovich et al). Women diagnosed and treated for breast cancer had higher biological aging metrics than women who were cancer-free at the time of follow-up: PhenoAgeAccel3 (standardized mean difference [β] = 0.13; P = .04), GrimAgeAccel4 (β = 0.14; P = .01), and DunedinPACE5 (β = 0.37; P < .001). Regarding breast cancer therapies received, the increases in biological aging were most striking for those women who underwent radiation. The effect of cancer treatments, specifically chemotherapy and radiation, on DNA methylation profiles and accelerating the aging process has been demonstrated in prior studies as well.6 Future research should strive to improve our understanding of the specific mechanisms underlying these age-related changes, identify ways to affect those which are modifiable, and positively influence long-term cognitive and functional consequences.

The association between cardiometabolic abnormalities, including obesity, hyperinsulinemia, diabetes, hypertension, and dyslipidemia, and an elevated breast cancer risk has been demonstrated in various studies.7 Furthermore, dysregulation of obesity-related proteins plays a role in breast cancer development and progression. A study by Xu and colleagues evaluated the temporal relationships and longitudinal associations of body mass index (BMI), cardiometabolic risk score (CRS), and obesity-related protein score (OPS) among 444 healthy women in a breast cancer screening cohort. After adjustment for demographics, lifestyle, and reproductive factors, a 1-kg/m2 increase in BMI per year increased CRS in both premenopausal (0.057 unit; P = .025) and postmenopausal women (0.054 unit; P = .033) and increased OPS by 0.588 unit (P = .001) in postmenopausal women. A significant association was also observed between CRS and OPS in postmenopausal women (β = 0.281; P = .034). These results support the importance of weight management and its effect on cardiometabolic and obesity-related parameters in breast cancer prevention. Research focused on lifestyle interventions to modify risk factors and effective implementation of these techniques will contribute to further reducing breast cancer risk.

Additional References

  1. García-Albéniz X, Hernán MA, Logan RW, et al. Continuation of annual screening mammography and breast cancer mortality in women older than 70 years. Ann Intern Med. 2020;172(6):381-389. doi: 10.7326/M18-1199
  2. Greenlee H, Iribarren C, Rana JS, et al. Risk of cardiovascular disease in women with and without breast cancer: The Pathways Heart Study. J Clin Oncol. 2022;40(15):1647-1658. doi: 10.1200/JCO.21.01736
  3. Levine ME, Lu AT, Quach A, et al. An epigenetic biomarker of aging for lifespan and healthspan. Aging (Albany NY). 2018;10(4):573-591. doi: 10.18632/aging.101414
  4. Lu AT, Quach A, Wilson JG, et al. DNA methylation GrimAge strongly predicts lifespan and healthspan. Aging (Albany NY). 2019;11(2):303-327. doi: 10.18632/aging.101684
  5. Belsky DW, Caspi A, Corcoran DL, et al. DunedinPACE, a DNA methylation biomarker of the pace of aging. eLife. 2022:11:e73420. doi: 10.7554/eLife.73420
  6. Sehl ME, Carroll JE, Horvath S, Bower JE. The acute effects of adjuvant radiation and chemotherapy on peripheral blood epigenetic age in early stage breast cancer patients. NPJ Breast Cancer. 2020;6:23. doi: 10.1038/s41523-020-0161-3
  7. Nouri M, Mohsenpour MA, Katsiki N, et al. Effect of serum lipid profile on the risk of breast cancer: Systematic review and meta-analysis of 1,628,871 women. J Clin Med. 2022;11(15):4503. doi: 10.3390/jcm11154503
Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Publications
Topics
Sections
Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Dr. Roesch scans the journals, so you don't have to!
Dr. Roesch scans the journals, so you don't have to!

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A11.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Overdiagnosis — defined as cancer detection, often through screening, that would not have led to symptoms during one’s lifetime — can be an issue associated with breast cancer screening in older women. Observational data have shown that continuing screening past age 75 years does not lead to substantial reductions in breast cancer mortality.1 A retrospective cohort study using the Surveillance, Epidemiology, and End Results (SEER)-Medicare registry, including 54,635 women ≥ 70 years of age, compared the cumulative incidence of breast cancer among older women who continued screening with those who did not and demonstrated that the relative risk for overdiagnosis increases with older age and lower life expectancy (Richman et al). The cumulative incidence of breast cancer was 6.1 vs 4.2 cases per 100 screened vs unscreened women among those aged 70-74 years, with an estimated 31% potentially overdiagnosed in the screened group. For women aged 75-84 years and ≥ 85 years, the estimated rates of breast cancer overdiagnosis were 47% and 54%, respectively. Furthermore, screening did not lead to a statistically significant difference in breast cancer–specific mortality in any of these age groups. The risks and benefits of breast cancer screening should be fully discussed with patients, and this decision-making process should consider a woman’s preference, comorbidities, and willingness to undergo specific treatments.

Studies have shown that breast cancer survivors have increased rates of age-related conditions, including cardiovascular disease and osteoporosis among others, therefore postulating that the biological aging process may be accelerated in this population.2 Among 417 women enrolled in the prospective Sister Study cohort, paired blood samples collected an average of 7.7 years apart compared three epigenetic metrics of biological aging (calculated on the basis of DNA methylation data) between women who were diagnosed and treated for breast cancer (n = 190) vs those who remained breast cancer–free (n = 227) (Kresovich et al). Women diagnosed and treated for breast cancer had higher biological aging metrics than women who were cancer-free at the time of follow-up: PhenoAgeAccel3 (standardized mean difference [β] = 0.13; P = .04), GrimAgeAccel4 (β = 0.14; P = .01), and DunedinPACE5 (β = 0.37; P < .001). Regarding breast cancer therapies received, the increases in biological aging were most striking for those women who underwent radiation. The effect of cancer treatments, specifically chemotherapy and radiation, on DNA methylation profiles and accelerating the aging process has been demonstrated in prior studies as well.6 Future research should strive to improve our understanding of the specific mechanisms underlying these age-related changes, identify ways to affect those which are modifiable, and positively influence long-term cognitive and functional consequences.

The association between cardiometabolic abnormalities, including obesity, hyperinsulinemia, diabetes, hypertension, and dyslipidemia, and an elevated breast cancer risk has been demonstrated in various studies.7 Furthermore, dysregulation of obesity-related proteins plays a role in breast cancer development and progression. A study by Xu and colleagues evaluated the temporal relationships and longitudinal associations of body mass index (BMI), cardiometabolic risk score (CRS), and obesity-related protein score (OPS) among 444 healthy women in a breast cancer screening cohort. After adjustment for demographics, lifestyle, and reproductive factors, a 1-kg/m2 increase in BMI per year increased CRS in both premenopausal (0.057 unit; P = .025) and postmenopausal women (0.054 unit; P = .033) and increased OPS by 0.588 unit (P = .001) in postmenopausal women. A significant association was also observed between CRS and OPS in postmenopausal women (β = 0.281; P = .034). These results support the importance of weight management and its effect on cardiometabolic and obesity-related parameters in breast cancer prevention. Research focused on lifestyle interventions to modify risk factors and effective implementation of these techniques will contribute to further reducing breast cancer risk.

Additional References

  1. García-Albéniz X, Hernán MA, Logan RW, et al. Continuation of annual screening mammography and breast cancer mortality in women older than 70 years. Ann Intern Med. 2020;172(6):381-389. doi: 10.7326/M18-1199
  2. Greenlee H, Iribarren C, Rana JS, et al. Risk of cardiovascular disease in women with and without breast cancer: The Pathways Heart Study. J Clin Oncol. 2022;40(15):1647-1658. doi: 10.1200/JCO.21.01736
  3. Levine ME, Lu AT, Quach A, et al. An epigenetic biomarker of aging for lifespan and healthspan. Aging (Albany NY). 2018;10(4):573-591. doi: 10.18632/aging.101414
  4. Lu AT, Quach A, Wilson JG, et al. DNA methylation GrimAge strongly predicts lifespan and healthspan. Aging (Albany NY). 2019;11(2):303-327. doi: 10.18632/aging.101684
  5. Belsky DW, Caspi A, Corcoran DL, et al. DunedinPACE, a DNA methylation biomarker of the pace of aging. eLife. 2022:11:e73420. doi: 10.7554/eLife.73420
  6. Sehl ME, Carroll JE, Horvath S, Bower JE. The acute effects of adjuvant radiation and chemotherapy on peripheral blood epigenetic age in early stage breast cancer patients. NPJ Breast Cancer. 2020;6:23. doi: 10.1038/s41523-020-0161-3
  7. Nouri M, Mohsenpour MA, Katsiki N, et al. Effect of serum lipid profile on the risk of breast cancer: Systematic review and meta-analysis of 1,628,871 women. J Clin Med. 2022;11(15):4503. doi: 10.3390/jcm11154503

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A11.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Overdiagnosis — defined as cancer detection, often through screening, that would not have led to symptoms during one’s lifetime — can be an issue associated with breast cancer screening in older women. Observational data have shown that continuing screening past age 75 years does not lead to substantial reductions in breast cancer mortality.1 A retrospective cohort study using the Surveillance, Epidemiology, and End Results (SEER)-Medicare registry, including 54,635 women ≥ 70 years of age, compared the cumulative incidence of breast cancer among older women who continued screening with those who did not and demonstrated that the relative risk for overdiagnosis increases with older age and lower life expectancy (Richman et al). The cumulative incidence of breast cancer was 6.1 vs 4.2 cases per 100 screened vs unscreened women among those aged 70-74 years, with an estimated 31% potentially overdiagnosed in the screened group. For women aged 75-84 years and ≥ 85 years, the estimated rates of breast cancer overdiagnosis were 47% and 54%, respectively. Furthermore, screening did not lead to a statistically significant difference in breast cancer–specific mortality in any of these age groups. The risks and benefits of breast cancer screening should be fully discussed with patients, and this decision-making process should consider a woman’s preference, comorbidities, and willingness to undergo specific treatments.

Studies have shown that breast cancer survivors have increased rates of age-related conditions, including cardiovascular disease and osteoporosis among others, therefore postulating that the biological aging process may be accelerated in this population.2 Among 417 women enrolled in the prospective Sister Study cohort, paired blood samples collected an average of 7.7 years apart compared three epigenetic metrics of biological aging (calculated on the basis of DNA methylation data) between women who were diagnosed and treated for breast cancer (n = 190) vs those who remained breast cancer–free (n = 227) (Kresovich et al). Women diagnosed and treated for breast cancer had higher biological aging metrics than women who were cancer-free at the time of follow-up: PhenoAgeAccel3 (standardized mean difference [β] = 0.13; P = .04), GrimAgeAccel4 (β = 0.14; P = .01), and DunedinPACE5 (β = 0.37; P < .001). Regarding breast cancer therapies received, the increases in biological aging were most striking for those women who underwent radiation. The effect of cancer treatments, specifically chemotherapy and radiation, on DNA methylation profiles and accelerating the aging process has been demonstrated in prior studies as well.6 Future research should strive to improve our understanding of the specific mechanisms underlying these age-related changes, identify ways to affect those which are modifiable, and positively influence long-term cognitive and functional consequences.

The association between cardiometabolic abnormalities, including obesity, hyperinsulinemia, diabetes, hypertension, and dyslipidemia, and an elevated breast cancer risk has been demonstrated in various studies.7 Furthermore, dysregulation of obesity-related proteins plays a role in breast cancer development and progression. A study by Xu and colleagues evaluated the temporal relationships and longitudinal associations of body mass index (BMI), cardiometabolic risk score (CRS), and obesity-related protein score (OPS) among 444 healthy women in a breast cancer screening cohort. After adjustment for demographics, lifestyle, and reproductive factors, a 1-kg/m2 increase in BMI per year increased CRS in both premenopausal (0.057 unit; P = .025) and postmenopausal women (0.054 unit; P = .033) and increased OPS by 0.588 unit (P = .001) in postmenopausal women. A significant association was also observed between CRS and OPS in postmenopausal women (β = 0.281; P = .034). These results support the importance of weight management and its effect on cardiometabolic and obesity-related parameters in breast cancer prevention. Research focused on lifestyle interventions to modify risk factors and effective implementation of these techniques will contribute to further reducing breast cancer risk.

Additional References

  1. García-Albéniz X, Hernán MA, Logan RW, et al. Continuation of annual screening mammography and breast cancer mortality in women older than 70 years. Ann Intern Med. 2020;172(6):381-389. doi: 10.7326/M18-1199
  2. Greenlee H, Iribarren C, Rana JS, et al. Risk of cardiovascular disease in women with and without breast cancer: The Pathways Heart Study. J Clin Oncol. 2022;40(15):1647-1658. doi: 10.1200/JCO.21.01736
  3. Levine ME, Lu AT, Quach A, et al. An epigenetic biomarker of aging for lifespan and healthspan. Aging (Albany NY). 2018;10(4):573-591. doi: 10.18632/aging.101414
  4. Lu AT, Quach A, Wilson JG, et al. DNA methylation GrimAge strongly predicts lifespan and healthspan. Aging (Albany NY). 2019;11(2):303-327. doi: 10.18632/aging.101684
  5. Belsky DW, Caspi A, Corcoran DL, et al. DunedinPACE, a DNA methylation biomarker of the pace of aging. eLife. 2022:11:e73420. doi: 10.7554/eLife.73420
  6. Sehl ME, Carroll JE, Horvath S, Bower JE. The acute effects of adjuvant radiation and chemotherapy on peripheral blood epigenetic age in early stage breast cancer patients. NPJ Breast Cancer. 2020;6:23. doi: 10.1038/s41523-020-0161-3
  7. Nouri M, Mohsenpour MA, Katsiki N, et al. Effect of serum lipid profile on the risk of breast cancer: Systematic review and meta-analysis of 1,628,871 women. J Clin Med. 2022;11(15):4503. doi: 10.3390/jcm11154503
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Mon, 05/03/2021 - 14:45
Un-Gate On Date
Mon, 05/03/2021 - 14:45
Use ProPublica
CFC Schedule Remove Status
Mon, 05/03/2021 - 14:45
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Activity Salesforce Deliverable ID
367005.2
Activity ID
93656
Product Name
Clinical Edge Journal Scan
Product ID
124
Supporter Name /ID
Perjeta [ 3532 ]

Commentary: Node irradiation, HER2+ treatment, and diet in BC, August 2023

Article Type
Changed
Fri, 08/04/2023 - 16:02
Dr. Roesch scans the journals, so you don't have to!

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A11.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Multiple previous trials have demonstrated the benefits of regional nodal irradiation (RNI) among patients with node-positive breast cancer, including postmastectomy and after breast-conserving surgery (BCS). The NCIC MA.20 trial, which included primarily patients with one to three involved nodes, demonstrated disease-free survival (DFS) improvement with the addition of RNI to whole-breast radiotherapy (DFS of 82.0% in the RNI group vs 77.0% in the control group; hazard ratio for DFS 0.76; P = .01).1 However, the selection of patients for RNI is variable and may depend on patient and tumor characteristics as well as surgery and the systemic therapies applied. In the NCIC MA.20 trial, nodal-irradiation was associated with better overall survival among those with estrogen receptor (ER)–negative breast cancer but not among those with ER-positive disease. A secondary analysis of the SWOG S1007 trial, which randomly assigned patients with hormone receptor (HR)–positive/human epidermal growth factor receptor 2 (HER2)–negative breast cancer with one to three involved nodes and a 21-gene recurrence score ≤ 25 to endocrine therapy alone or chemotherapy plus endocrine therapy, investigated the use of radiotherapy and patterns of locoregional recurrence (Jagsi et al). Of those patients who received radiotherapy with complete information on targets (N = 3852), 59% (N = 2274) received RNI. At median follow-up of 6.1 years, the cumulative incidence of locoregional recurrence was low among all groups: 0.85% after BCS and radiotherapy with RNI, 0.55% after BCS with radiotherapy without RNI, 0.11% after mastectomy with postmastectomy radiation therapy (PMRT), and 1.7% after mastectomy without radiotherapy. Receiving RNI was not associated with invasive DFS for pre- or postmenopausal patients. These data support the importance of prospective studies, including the NCIC MA.39 trial,2 designed to identify optimal locoregional therapy in patients with limited nodal burden and favorable disease biology.

The addition of pertuzumab to trastuzumab plus chemotherapy has demonstrated improvement in pathologic complete response (pCR) rates compared with trastuzumab plus chemotherapy in early-stage HER2-positive breast cancer.3 The framework of oncology is built on clinical trials through their rigorous design, enrollment, and synthesis of data; however, real-world studies are an integral component of cancer research because they provide a more representative sample of the general population treated in routine clinical practice. Neopearl was a retrospective, observational, real-world study that evaluated the efficacy and safety of trastuzumab plus chemotherapy with or without pertuzumab among 271 patients with stage II-III HER2-positive breast cancer (Fabbri et al). The addition of pertuzumab led to an increase in pCR rate (49% vs 62%; odds ratio 1.74; P = .032) and improvement in 5-year event-free survival (81% vs 93%; hazard ratio 2.22; P = .041), and the benefit on univariate analysis was restricted to patients with positive axillary nodes. Furthermore, there were no significant differences in adverse events, including cardiac, between the two groups. These results serve to strengthen the available data regarding the clinical efficacy and favorable safety profile of dual HER2-targeted therapy combined with neoadjuvant chemotherapy.

Lifestyle factors, including physical activity and diet, are becoming increasingly recognized as important determinants of various cancer-specific outcomes and overall health. Furthermore, because these are modifiable, there is often motivation on behalf of an individual to change behaviors that can affect their outcome. Adherence to the Mediterranean diet (MD) has been associated with reduced risk for breast cancer development and lower mortality among women with breast cancer.4,5 Data from a prospective multicenter European cohort including 13,270 breast cancer survivors demonstrated that low compared with medium adherence to a MD before a breast cancer diagnosis was associated with a 13% higher risk for all-cause mortality (hazard ratio 1.13; 95% CI 1.01-1.26). A three-unit increase in the adapted relative MD score was associated with an 8% reduced risk for overall mortality (hazard ratio3-unit 0.92; 95% CI 0.87-0.97); this result was sustained in the postmenopausal population and strengthened in metastatic disease (Castro-Espin et al). The connection between diet and cancer outcomes is complex, and future research evaluating specific dietary interventions and the underlying biologic pathways by which nutrition exerts its effects will be important to inform our counseling for patients with breast cancer in the survivorship setting.

Additional References

  1. Whelan TJ, Olivotto IA, Parulekar WR, et al, for the MA.20 Study Investigators. Regional nodal irradiation in early-stage breast cancer. N Engl J Med. 2015;373:307-16. doi:10.1056/NEJMoa1415340
  2. ClinicalTrials.gov. Regional radiotherapy in biomarker low-risk node positive and T3N0 breast cancer (TAILOR RT). National Library of Medicine. Last updated November 23, 2022. https://www.clinicaltrials.gov/study/NCT03488693
  3. Gianni L, Pienkowski T, Im YH, et al. Efficacy and safety of neoadjuvant pertuzumab and trastuzumab in women with locally advanced, inflammatory, or early HER2-positive breast cancer (NeoSphere): A randomised multicentre, open-label, phase 2 trial. Lancet Oncol. 2012;13:25-32. doi:10.1016/S1470-2045(11)70336-9
  4. Buckland G, Travier N, Cottet V, et al. Adherence to the mediterranean diet and risk of breast cancer in the European prospective investigation into cancer and nutrition cohort study. Int J Cancer. 2013;132:2918-27. doi:10.1002/ijc.27958
  5. Haslam DE, John EM, Knight JA, et al. Diet quality and all-cause mortality in women with breast cancer from the Breast Cancer Family Registry. Cancer Epidemiol Biomarkers Prev. 2023;32:678-686. doi:10.1158/1055-9965.EPI-22-1198
Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Publications
Topics
Sections
Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Dr. Roesch scans the journals, so you don't have to!
Dr. Roesch scans the journals, so you don't have to!

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A11.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Multiple previous trials have demonstrated the benefits of regional nodal irradiation (RNI) among patients with node-positive breast cancer, including postmastectomy and after breast-conserving surgery (BCS). The NCIC MA.20 trial, which included primarily patients with one to three involved nodes, demonstrated disease-free survival (DFS) improvement with the addition of RNI to whole-breast radiotherapy (DFS of 82.0% in the RNI group vs 77.0% in the control group; hazard ratio for DFS 0.76; P = .01).1 However, the selection of patients for RNI is variable and may depend on patient and tumor characteristics as well as surgery and the systemic therapies applied. In the NCIC MA.20 trial, nodal-irradiation was associated with better overall survival among those with estrogen receptor (ER)–negative breast cancer but not among those with ER-positive disease. A secondary analysis of the SWOG S1007 trial, which randomly assigned patients with hormone receptor (HR)–positive/human epidermal growth factor receptor 2 (HER2)–negative breast cancer with one to three involved nodes and a 21-gene recurrence score ≤ 25 to endocrine therapy alone or chemotherapy plus endocrine therapy, investigated the use of radiotherapy and patterns of locoregional recurrence (Jagsi et al). Of those patients who received radiotherapy with complete information on targets (N = 3852), 59% (N = 2274) received RNI. At median follow-up of 6.1 years, the cumulative incidence of locoregional recurrence was low among all groups: 0.85% after BCS and radiotherapy with RNI, 0.55% after BCS with radiotherapy without RNI, 0.11% after mastectomy with postmastectomy radiation therapy (PMRT), and 1.7% after mastectomy without radiotherapy. Receiving RNI was not associated with invasive DFS for pre- or postmenopausal patients. These data support the importance of prospective studies, including the NCIC MA.39 trial,2 designed to identify optimal locoregional therapy in patients with limited nodal burden and favorable disease biology.

The addition of pertuzumab to trastuzumab plus chemotherapy has demonstrated improvement in pathologic complete response (pCR) rates compared with trastuzumab plus chemotherapy in early-stage HER2-positive breast cancer.3 The framework of oncology is built on clinical trials through their rigorous design, enrollment, and synthesis of data; however, real-world studies are an integral component of cancer research because they provide a more representative sample of the general population treated in routine clinical practice. Neopearl was a retrospective, observational, real-world study that evaluated the efficacy and safety of trastuzumab plus chemotherapy with or without pertuzumab among 271 patients with stage II-III HER2-positive breast cancer (Fabbri et al). The addition of pertuzumab led to an increase in pCR rate (49% vs 62%; odds ratio 1.74; P = .032) and improvement in 5-year event-free survival (81% vs 93%; hazard ratio 2.22; P = .041), and the benefit on univariate analysis was restricted to patients with positive axillary nodes. Furthermore, there were no significant differences in adverse events, including cardiac, between the two groups. These results serve to strengthen the available data regarding the clinical efficacy and favorable safety profile of dual HER2-targeted therapy combined with neoadjuvant chemotherapy.

Lifestyle factors, including physical activity and diet, are becoming increasingly recognized as important determinants of various cancer-specific outcomes and overall health. Furthermore, because these are modifiable, there is often motivation on behalf of an individual to change behaviors that can affect their outcome. Adherence to the Mediterranean diet (MD) has been associated with reduced risk for breast cancer development and lower mortality among women with breast cancer.4,5 Data from a prospective multicenter European cohort including 13,270 breast cancer survivors demonstrated that low compared with medium adherence to a MD before a breast cancer diagnosis was associated with a 13% higher risk for all-cause mortality (hazard ratio 1.13; 95% CI 1.01-1.26). A three-unit increase in the adapted relative MD score was associated with an 8% reduced risk for overall mortality (hazard ratio3-unit 0.92; 95% CI 0.87-0.97); this result was sustained in the postmenopausal population and strengthened in metastatic disease (Castro-Espin et al). The connection between diet and cancer outcomes is complex, and future research evaluating specific dietary interventions and the underlying biologic pathways by which nutrition exerts its effects will be important to inform our counseling for patients with breast cancer in the survivorship setting.

Additional References

  1. Whelan TJ, Olivotto IA, Parulekar WR, et al, for the MA.20 Study Investigators. Regional nodal irradiation in early-stage breast cancer. N Engl J Med. 2015;373:307-16. doi:10.1056/NEJMoa1415340
  2. ClinicalTrials.gov. Regional radiotherapy in biomarker low-risk node positive and T3N0 breast cancer (TAILOR RT). National Library of Medicine. Last updated November 23, 2022. https://www.clinicaltrials.gov/study/NCT03488693
  3. Gianni L, Pienkowski T, Im YH, et al. Efficacy and safety of neoadjuvant pertuzumab and trastuzumab in women with locally advanced, inflammatory, or early HER2-positive breast cancer (NeoSphere): A randomised multicentre, open-label, phase 2 trial. Lancet Oncol. 2012;13:25-32. doi:10.1016/S1470-2045(11)70336-9
  4. Buckland G, Travier N, Cottet V, et al. Adherence to the mediterranean diet and risk of breast cancer in the European prospective investigation into cancer and nutrition cohort study. Int J Cancer. 2013;132:2918-27. doi:10.1002/ijc.27958
  5. Haslam DE, John EM, Knight JA, et al. Diet quality and all-cause mortality in women with breast cancer from the Breast Cancer Family Registry. Cancer Epidemiol Biomarkers Prev. 2023;32:678-686. doi:10.1158/1055-9965.EPI-22-1198

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A11.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Multiple previous trials have demonstrated the benefits of regional nodal irradiation (RNI) among patients with node-positive breast cancer, including postmastectomy and after breast-conserving surgery (BCS). The NCIC MA.20 trial, which included primarily patients with one to three involved nodes, demonstrated disease-free survival (DFS) improvement with the addition of RNI to whole-breast radiotherapy (DFS of 82.0% in the RNI group vs 77.0% in the control group; hazard ratio for DFS 0.76; P = .01).1 However, the selection of patients for RNI is variable and may depend on patient and tumor characteristics as well as surgery and the systemic therapies applied. In the NCIC MA.20 trial, nodal-irradiation was associated with better overall survival among those with estrogen receptor (ER)–negative breast cancer but not among those with ER-positive disease. A secondary analysis of the SWOG S1007 trial, which randomly assigned patients with hormone receptor (HR)–positive/human epidermal growth factor receptor 2 (HER2)–negative breast cancer with one to three involved nodes and a 21-gene recurrence score ≤ 25 to endocrine therapy alone or chemotherapy plus endocrine therapy, investigated the use of radiotherapy and patterns of locoregional recurrence (Jagsi et al). Of those patients who received radiotherapy with complete information on targets (N = 3852), 59% (N = 2274) received RNI. At median follow-up of 6.1 years, the cumulative incidence of locoregional recurrence was low among all groups: 0.85% after BCS and radiotherapy with RNI, 0.55% after BCS with radiotherapy without RNI, 0.11% after mastectomy with postmastectomy radiation therapy (PMRT), and 1.7% after mastectomy without radiotherapy. Receiving RNI was not associated with invasive DFS for pre- or postmenopausal patients. These data support the importance of prospective studies, including the NCIC MA.39 trial,2 designed to identify optimal locoregional therapy in patients with limited nodal burden and favorable disease biology.

The addition of pertuzumab to trastuzumab plus chemotherapy has demonstrated improvement in pathologic complete response (pCR) rates compared with trastuzumab plus chemotherapy in early-stage HER2-positive breast cancer.3 The framework of oncology is built on clinical trials through their rigorous design, enrollment, and synthesis of data; however, real-world studies are an integral component of cancer research because they provide a more representative sample of the general population treated in routine clinical practice. Neopearl was a retrospective, observational, real-world study that evaluated the efficacy and safety of trastuzumab plus chemotherapy with or without pertuzumab among 271 patients with stage II-III HER2-positive breast cancer (Fabbri et al). The addition of pertuzumab led to an increase in pCR rate (49% vs 62%; odds ratio 1.74; P = .032) and improvement in 5-year event-free survival (81% vs 93%; hazard ratio 2.22; P = .041), and the benefit on univariate analysis was restricted to patients with positive axillary nodes. Furthermore, there were no significant differences in adverse events, including cardiac, between the two groups. These results serve to strengthen the available data regarding the clinical efficacy and favorable safety profile of dual HER2-targeted therapy combined with neoadjuvant chemotherapy.

Lifestyle factors, including physical activity and diet, are becoming increasingly recognized as important determinants of various cancer-specific outcomes and overall health. Furthermore, because these are modifiable, there is often motivation on behalf of an individual to change behaviors that can affect their outcome. Adherence to the Mediterranean diet (MD) has been associated with reduced risk for breast cancer development and lower mortality among women with breast cancer.4,5 Data from a prospective multicenter European cohort including 13,270 breast cancer survivors demonstrated that low compared with medium adherence to a MD before a breast cancer diagnosis was associated with a 13% higher risk for all-cause mortality (hazard ratio 1.13; 95% CI 1.01-1.26). A three-unit increase in the adapted relative MD score was associated with an 8% reduced risk for overall mortality (hazard ratio3-unit 0.92; 95% CI 0.87-0.97); this result was sustained in the postmenopausal population and strengthened in metastatic disease (Castro-Espin et al). The connection between diet and cancer outcomes is complex, and future research evaluating specific dietary interventions and the underlying biologic pathways by which nutrition exerts its effects will be important to inform our counseling for patients with breast cancer in the survivorship setting.

Additional References

  1. Whelan TJ, Olivotto IA, Parulekar WR, et al, for the MA.20 Study Investigators. Regional nodal irradiation in early-stage breast cancer. N Engl J Med. 2015;373:307-16. doi:10.1056/NEJMoa1415340
  2. ClinicalTrials.gov. Regional radiotherapy in biomarker low-risk node positive and T3N0 breast cancer (TAILOR RT). National Library of Medicine. Last updated November 23, 2022. https://www.clinicaltrials.gov/study/NCT03488693
  3. Gianni L, Pienkowski T, Im YH, et al. Efficacy and safety of neoadjuvant pertuzumab and trastuzumab in women with locally advanced, inflammatory, or early HER2-positive breast cancer (NeoSphere): A randomised multicentre, open-label, phase 2 trial. Lancet Oncol. 2012;13:25-32. doi:10.1016/S1470-2045(11)70336-9
  4. Buckland G, Travier N, Cottet V, et al. Adherence to the mediterranean diet and risk of breast cancer in the European prospective investigation into cancer and nutrition cohort study. Int J Cancer. 2013;132:2918-27. doi:10.1002/ijc.27958
  5. Haslam DE, John EM, Knight JA, et al. Diet quality and all-cause mortality in women with breast cancer from the Breast Cancer Family Registry. Cancer Epidemiol Biomarkers Prev. 2023;32:678-686. doi:10.1158/1055-9965.EPI-22-1198
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Mon, 05/03/2021 - 14:45
Un-Gate On Date
Mon, 05/03/2021 - 14:45
Use ProPublica
CFC Schedule Remove Status
Mon, 05/03/2021 - 14:45
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Activity Salesforce Deliverable ID
367005.2
Activity ID
93656
Product Name
Clinical Edge Journal Scan
Product ID
124
Supporter Name /ID
Perjeta [ 3532 ]

Commentary: Advances in HER2 advanced breast cancer, July 2023

Article Type
Changed
Wed, 07/05/2023 - 11:45
Dr. Roesch scans the journals, so you don't have to!

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Alterations in the PI3K/AKT/PTEN pathway are present in about half of hormone receptor–positive (HR+)/human epidermal growth factor 2–negative (HER2-) breast cancers and may mediate endocrine resistance in this population. The phase 2 BYLieve trial demonstrated activity of alpelisib + fulvestrant in patients with PIK3CA-mutated, HR+/HER2- advanced breast cancer (ABC) in the post–CDK4/6 inhibitor setting.1 Capivasertib, an oral selective inhibitor of all three AKT isoforms, was investigated in the phase 3 CAPItello-291 trial among 708 patients with HR+/HER2- ABC who had relapsed or had disease progression on or after aromatase inhibitor therapy with or without a CDK4/6 inhibitor. The combination of capivasertib + fulvestrant led to a significant improvement in progression-free survival (PFS) vs placebo + fulvestrant both in the overall population (median PFS 7.2 mo vs 3.6 mo; hazard ratio 0.60; P < .001) and in patients with AKT pathway-altered (PIK3CA, AKT1, or PTEN) tumors (median PFS 7.3 mo vs 3.1 mo; hazard ratio 0.50; P < .001) (Turner et al). The most common grade 3 or higher adverse events were rash and diarrhea in the capivasertib + fulvestrant arm (12.1% and 9.3%, respectively), and the discontinuation rate was 13.0%. These results highlight the activity of this combination and its overall acceptable safety profile and present a potential new therapy option for patients. The selection, optimal sequencing, and combinations of new agents in the HR+/HER2 ABC space remains an area of active research.

The neoadjuvant setting provides a favorable environment to study de-escalation approaches as treatment response (via pathologic complete response [pCR] assessment) can be used as a surrogate marker for outcome. Studies have shown the effect of HER2-enriched subtype and high ERBB2 expression on pCR rates after receipt of a chemotherapy-free, dual HER2-targeted regimen.2 The prospective, multicenter, neoadjuvant phase 2 WSG-TP-II trial randomly assigned 207 patients with HR+/HER2+ early breast cancer to 12 weeks of endocrine therapy (ET)–trastuzumab-pertuzumab vs paclitaxel-trastuzumab-pertuzumab. The pCR rate was inferior in the ET arm compared with the paclitaxel arm (23.7% vs 56.4%; odds ratio 0.24; 95% CI 0.12-0.46; P < .001). In addition, an immunohistochemistry ERBB2 score of 3 or higher and ERBB2-enriched subtype were predictors of higher pCR rates in both arms (Gluz et al). This study not only supports a deescalated chemotherapy neoadjuvant strategy of paclitaxel + dual HER2 blockade but also suggests that a portion of patients may potentially be spared chemotherapy with very good results. The role of biomarkers is integral to patient selection for these approaches, and the evaluation of response in real-time will allow for the tailoring of therapy to achieve the best outcome.

Systemic staging for locally advanced breast cancer (LABC) is important for informing prognosis as well as aiding in development of an appropriate treatment plan for patients. The PETABC study included 369 patients with LABC (TNM stage III or IIB [T3N0]) with random assignment to 18F-labeled fluorodeoxyglucose PET-CT or conventional staging (bone scan, CT of chest/abdomen/pelvis), and was designed to assess the rate of upstaging with each imaging modality and effect on treatment (Dayes et al). In the PET-CT group, 23% (N = 43) of patients were upstaged to stage IV compared with 11% (N = 21) in the conventional-staging group (absolute difference 12.3%; 95% CI 3.9-19.9; P = .002). Fewer patients in the PET-CT group received combined modality treatment vs those patients in the conventional staging group (81% vs 89.2%; P = .03). These results support the consideration of PET-CT as a staging tool for LABC, and this is reflected in various clinical guidelines. Furthermore, the evolving role of other imaging techniques such as 18F-fluoroestradiol (18F-FES) PET-CT in detection of metastatic lesions related to estrogen receptor–positive breast cancer3 will continue to advance the field of imaging.

Additional References

  1. Rugo HS, Lerebours F, Ciruelos E, et al. Alpelisib plus fulvestrant in PIK3CA-mutated, hormone receptor-positive advanced breast cancer after a CDK4/6 inhibitor (BYLieve): One cohort of a phase 2, multicentre, open-label, non-comparative study. Lancet Oncol. 2021;22:489-498. doi: 10.1016/S1470-2045(21)00034-6. Erratum in: Lancet Oncol. 2021;22(5):e184. doi: 10.1016/S1470-2045(21)00194-7
  2. Prat A, Pascual T, De Angelis C, et al. HER2-enriched subtype and ERBB2 expression in HER2-positive breast cancer treated with dual HER2 blockade. J Natl Cancer Inst. 2020;112:46-54. doi: 10.1093/jnci/djz042
  3. Ulaner GA, Jhaveri K, Chandarlapaty S, et al. Head-to-head evaluation of 18F-FES and 18F-FDG PET/CT in metastatic invasive lobular breast cancer. J Nucl Med. 2021;62:326-331. doi: 10.2967/jnumed.120.247882
Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Publications
Topics
Sections
Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Dr. Roesch scans the journals, so you don't have to!
Dr. Roesch scans the journals, so you don't have to!

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Alterations in the PI3K/AKT/PTEN pathway are present in about half of hormone receptor–positive (HR+)/human epidermal growth factor 2–negative (HER2-) breast cancers and may mediate endocrine resistance in this population. The phase 2 BYLieve trial demonstrated activity of alpelisib + fulvestrant in patients with PIK3CA-mutated, HR+/HER2- advanced breast cancer (ABC) in the post–CDK4/6 inhibitor setting.1 Capivasertib, an oral selective inhibitor of all three AKT isoforms, was investigated in the phase 3 CAPItello-291 trial among 708 patients with HR+/HER2- ABC who had relapsed or had disease progression on or after aromatase inhibitor therapy with or without a CDK4/6 inhibitor. The combination of capivasertib + fulvestrant led to a significant improvement in progression-free survival (PFS) vs placebo + fulvestrant both in the overall population (median PFS 7.2 mo vs 3.6 mo; hazard ratio 0.60; P < .001) and in patients with AKT pathway-altered (PIK3CA, AKT1, or PTEN) tumors (median PFS 7.3 mo vs 3.1 mo; hazard ratio 0.50; P < .001) (Turner et al). The most common grade 3 or higher adverse events were rash and diarrhea in the capivasertib + fulvestrant arm (12.1% and 9.3%, respectively), and the discontinuation rate was 13.0%. These results highlight the activity of this combination and its overall acceptable safety profile and present a potential new therapy option for patients. The selection, optimal sequencing, and combinations of new agents in the HR+/HER2 ABC space remains an area of active research.

The neoadjuvant setting provides a favorable environment to study de-escalation approaches as treatment response (via pathologic complete response [pCR] assessment) can be used as a surrogate marker for outcome. Studies have shown the effect of HER2-enriched subtype and high ERBB2 expression on pCR rates after receipt of a chemotherapy-free, dual HER2-targeted regimen.2 The prospective, multicenter, neoadjuvant phase 2 WSG-TP-II trial randomly assigned 207 patients with HR+/HER2+ early breast cancer to 12 weeks of endocrine therapy (ET)–trastuzumab-pertuzumab vs paclitaxel-trastuzumab-pertuzumab. The pCR rate was inferior in the ET arm compared with the paclitaxel arm (23.7% vs 56.4%; odds ratio 0.24; 95% CI 0.12-0.46; P < .001). In addition, an immunohistochemistry ERBB2 score of 3 or higher and ERBB2-enriched subtype were predictors of higher pCR rates in both arms (Gluz et al). This study not only supports a deescalated chemotherapy neoadjuvant strategy of paclitaxel + dual HER2 blockade but also suggests that a portion of patients may potentially be spared chemotherapy with very good results. The role of biomarkers is integral to patient selection for these approaches, and the evaluation of response in real-time will allow for the tailoring of therapy to achieve the best outcome.

Systemic staging for locally advanced breast cancer (LABC) is important for informing prognosis as well as aiding in development of an appropriate treatment plan for patients. The PETABC study included 369 patients with LABC (TNM stage III or IIB [T3N0]) with random assignment to 18F-labeled fluorodeoxyglucose PET-CT or conventional staging (bone scan, CT of chest/abdomen/pelvis), and was designed to assess the rate of upstaging with each imaging modality and effect on treatment (Dayes et al). In the PET-CT group, 23% (N = 43) of patients were upstaged to stage IV compared with 11% (N = 21) in the conventional-staging group (absolute difference 12.3%; 95% CI 3.9-19.9; P = .002). Fewer patients in the PET-CT group received combined modality treatment vs those patients in the conventional staging group (81% vs 89.2%; P = .03). These results support the consideration of PET-CT as a staging tool for LABC, and this is reflected in various clinical guidelines. Furthermore, the evolving role of other imaging techniques such as 18F-fluoroestradiol (18F-FES) PET-CT in detection of metastatic lesions related to estrogen receptor–positive breast cancer3 will continue to advance the field of imaging.

Additional References

  1. Rugo HS, Lerebours F, Ciruelos E, et al. Alpelisib plus fulvestrant in PIK3CA-mutated, hormone receptor-positive advanced breast cancer after a CDK4/6 inhibitor (BYLieve): One cohort of a phase 2, multicentre, open-label, non-comparative study. Lancet Oncol. 2021;22:489-498. doi: 10.1016/S1470-2045(21)00034-6. Erratum in: Lancet Oncol. 2021;22(5):e184. doi: 10.1016/S1470-2045(21)00194-7
  2. Prat A, Pascual T, De Angelis C, et al. HER2-enriched subtype and ERBB2 expression in HER2-positive breast cancer treated with dual HER2 blockade. J Natl Cancer Inst. 2020;112:46-54. doi: 10.1093/jnci/djz042
  3. Ulaner GA, Jhaveri K, Chandarlapaty S, et al. Head-to-head evaluation of 18F-FES and 18F-FDG PET/CT in metastatic invasive lobular breast cancer. J Nucl Med. 2021;62:326-331. doi: 10.2967/jnumed.120.247882

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Alterations in the PI3K/AKT/PTEN pathway are present in about half of hormone receptor–positive (HR+)/human epidermal growth factor 2–negative (HER2-) breast cancers and may mediate endocrine resistance in this population. The phase 2 BYLieve trial demonstrated activity of alpelisib + fulvestrant in patients with PIK3CA-mutated, HR+/HER2- advanced breast cancer (ABC) in the post–CDK4/6 inhibitor setting.1 Capivasertib, an oral selective inhibitor of all three AKT isoforms, was investigated in the phase 3 CAPItello-291 trial among 708 patients with HR+/HER2- ABC who had relapsed or had disease progression on or after aromatase inhibitor therapy with or without a CDK4/6 inhibitor. The combination of capivasertib + fulvestrant led to a significant improvement in progression-free survival (PFS) vs placebo + fulvestrant both in the overall population (median PFS 7.2 mo vs 3.6 mo; hazard ratio 0.60; P < .001) and in patients with AKT pathway-altered (PIK3CA, AKT1, or PTEN) tumors (median PFS 7.3 mo vs 3.1 mo; hazard ratio 0.50; P < .001) (Turner et al). The most common grade 3 or higher adverse events were rash and diarrhea in the capivasertib + fulvestrant arm (12.1% and 9.3%, respectively), and the discontinuation rate was 13.0%. These results highlight the activity of this combination and its overall acceptable safety profile and present a potential new therapy option for patients. The selection, optimal sequencing, and combinations of new agents in the HR+/HER2 ABC space remains an area of active research.

The neoadjuvant setting provides a favorable environment to study de-escalation approaches as treatment response (via pathologic complete response [pCR] assessment) can be used as a surrogate marker for outcome. Studies have shown the effect of HER2-enriched subtype and high ERBB2 expression on pCR rates after receipt of a chemotherapy-free, dual HER2-targeted regimen.2 The prospective, multicenter, neoadjuvant phase 2 WSG-TP-II trial randomly assigned 207 patients with HR+/HER2+ early breast cancer to 12 weeks of endocrine therapy (ET)–trastuzumab-pertuzumab vs paclitaxel-trastuzumab-pertuzumab. The pCR rate was inferior in the ET arm compared with the paclitaxel arm (23.7% vs 56.4%; odds ratio 0.24; 95% CI 0.12-0.46; P < .001). In addition, an immunohistochemistry ERBB2 score of 3 or higher and ERBB2-enriched subtype were predictors of higher pCR rates in both arms (Gluz et al). This study not only supports a deescalated chemotherapy neoadjuvant strategy of paclitaxel + dual HER2 blockade but also suggests that a portion of patients may potentially be spared chemotherapy with very good results. The role of biomarkers is integral to patient selection for these approaches, and the evaluation of response in real-time will allow for the tailoring of therapy to achieve the best outcome.

Systemic staging for locally advanced breast cancer (LABC) is important for informing prognosis as well as aiding in development of an appropriate treatment plan for patients. The PETABC study included 369 patients with LABC (TNM stage III or IIB [T3N0]) with random assignment to 18F-labeled fluorodeoxyglucose PET-CT or conventional staging (bone scan, CT of chest/abdomen/pelvis), and was designed to assess the rate of upstaging with each imaging modality and effect on treatment (Dayes et al). In the PET-CT group, 23% (N = 43) of patients were upstaged to stage IV compared with 11% (N = 21) in the conventional-staging group (absolute difference 12.3%; 95% CI 3.9-19.9; P = .002). Fewer patients in the PET-CT group received combined modality treatment vs those patients in the conventional staging group (81% vs 89.2%; P = .03). These results support the consideration of PET-CT as a staging tool for LABC, and this is reflected in various clinical guidelines. Furthermore, the evolving role of other imaging techniques such as 18F-fluoroestradiol (18F-FES) PET-CT in detection of metastatic lesions related to estrogen receptor–positive breast cancer3 will continue to advance the field of imaging.

Additional References

  1. Rugo HS, Lerebours F, Ciruelos E, et al. Alpelisib plus fulvestrant in PIK3CA-mutated, hormone receptor-positive advanced breast cancer after a CDK4/6 inhibitor (BYLieve): One cohort of a phase 2, multicentre, open-label, non-comparative study. Lancet Oncol. 2021;22:489-498. doi: 10.1016/S1470-2045(21)00034-6. Erratum in: Lancet Oncol. 2021;22(5):e184. doi: 10.1016/S1470-2045(21)00194-7
  2. Prat A, Pascual T, De Angelis C, et al. HER2-enriched subtype and ERBB2 expression in HER2-positive breast cancer treated with dual HER2 blockade. J Natl Cancer Inst. 2020;112:46-54. doi: 10.1093/jnci/djz042
  3. Ulaner GA, Jhaveri K, Chandarlapaty S, et al. Head-to-head evaluation of 18F-FES and 18F-FDG PET/CT in metastatic invasive lobular breast cancer. J Nucl Med. 2021;62:326-331. doi: 10.2967/jnumed.120.247882
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Mon, 05/03/2021 - 14:45
Un-Gate On Date
Mon, 05/03/2021 - 14:45
Use ProPublica
CFC Schedule Remove Status
Mon, 05/03/2021 - 14:45
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Activity Salesforce Deliverable ID
367005.2
Activity ID
93656
Product Name
Clinical Edge Journal Scan
Product ID
124
Supporter Name /ID
Perjeta [ 3532 ]