1
|
Khan S, Jandrajupalli SB, Bushara NZA, Raja RDP, Mirza S, Sharma K, Verma R, Kumar A, Lohani M. Targeting Refractory Triple-Negative Breast Cancer with Sacituzumab Govitecan: A New Era in Precision Medicine. Cells 2024; 13:2126. [PMID: 39768216 PMCID: PMC11674573 DOI: 10.3390/cells13242126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/13/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Advanced triple-negative breast cancer (TNBC) has poorer outcomes due to its aggressive behavior and restricted therapeutic options. While therapies like checkpoint inhibitors and PARP inhibitors offer some benefits, chemotherapy remains ineffective beyond the first line of treatment. Antibody-drug conjugates (ADCs) like sacituzumab govitecan-hziy (SG) represent a significant advancement. SG combines SN-38, an irinotecan derivative, with a Trop-2-targeting antibody via a pH-sensitive linking moiety, achieving a good drug:antibody ratio. In a phase I-II study involving metastatic TNBC (mTNBC) individuals, SG achieved an overall response rate of 33.3% and a median response period of 7.7 months. The phase III ASCENT trial demonstrated SG's efficacy in relapsed or refractory TNBC, improving median progression-free survival and median overall survival compared to chemotherapy. Common side effects include neutropenia, nausea, and fatigue. This article highlights the clinical potential, pharmacokinetics, safety profile, and resistance mechanisms of SG along with key ongoing clinical trials, emphasizing its role in managing refractory mTNBC, especially in third-line therapy. The review also discusses current strategies for managing adverse reactions and sequencing ADC treatments in clinical practice, along with the predicted basis of resistance. The optimal sequencing of SG relative to other ADCs, such as trastuzumab deruxtecan or T-DXd, remains an evolving question, especially as newer agents with distinct mechanisms of action and safety profiles enter the field. Further research is essential to establish evidence-based strategies for sequencing SG and addressing disease progression post-ADC therapy.
Collapse
Affiliation(s)
- Saif Khan
- Department of Basic Dental and Medical Sciences, College of Dentistry, University of Ha’il, Ha’il 55473, Saudi Arabia; (S.K.); (S.M.)
| | - Suresh Babu Jandrajupalli
- Department of Preventive Dental Sciences, College of Dentistry, University of Ha’il, Ha’il 55473, Saudi Arabia; (S.B.J.); (N.Z.A.B.)
| | - Nashwa Zaki Ali Bushara
- Department of Preventive Dental Sciences, College of Dentistry, University of Ha’il, Ha’il 55473, Saudi Arabia; (S.B.J.); (N.Z.A.B.)
| | - Rama Devi Patel Raja
- Department of Biology, College of Science, University of Ha’il, Ha’il 55473, Saudi Arabia;
| | - Shadab Mirza
- Department of Basic Dental and Medical Sciences, College of Dentistry, University of Ha’il, Ha’il 55473, Saudi Arabia; (S.K.); (S.M.)
| | - Kuldeep Sharma
- Centre of Research Impact and Outcome, Chitkara University, Rajpura 140401, India;
| | - Rajan Verma
- Chitkara Center for Research and Development, Chitkara University, Baddi 174103, India;
| | - Ashish Kumar
- Department of Mechanical Engineering, Institute of Aeronautical Engineering, Hyderabad 500043, India;
- Division of Research and Development, Lovely Professional University, Phagwara 144411, India
| | - Mohtashim Lohani
- Department of Nursing, College of Nursing and Health Sciences, Jazan University, Jazan 45142, Saudi Arabia
| |
Collapse
|
2
|
Tao X, Han J, Li Y, Tian Y, Li ZJ, Li J, Guo X, Zhao J. The Difference of RCB 0 and RCB I in Prognosis of Breast Cancer After Neoadjuvant Therapy: A Meta-Analysis. Clin Breast Cancer 2024:S1526-8209(24)00336-7. [PMID: 39721893 DOI: 10.1016/j.clbc.2024.11.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/15/2024] [Accepted: 11/28/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND The use of the residual cancer burden (RCB) for assessing breast cancer after neoadjuvant therapy (NAT) is increasingly common, but the prognostic difference between RCB 0 and RCB I is unclear. METHODS We systematically reviewed literature from PubMed, Embase, Web of Science, and oncology conferences until September 24, 2023. We used fixed- and random-effects models to calculate hazard ratio (HR) with 95% confidence interval (CI) for event-free survival (EFS), overall survival (OS), and distant disease-free survival (DDFS). RESULTS Our meta-analysis, encompassing 19 studies with 5894 patients, revealed that in the general population, RCB I had worse EFS (HR = 2.13; 95% CI: 1.75-2.58), OS (HR = 2.08; 95% CI: 1.48-2.93), and DDFS (HR = 2.10; 95% CI: 1.65-2.67) than RCB 0. Consistent with results from the general population, RCB I exhibited poorer EFS, OS, and DDFS in human epidermal growth factor 2-positive (HER2+) subtype and triple-negative breast cancer (TNBC) compared to RCB 0. Conversely, luminal subtype with RCB 0 and RCB I showed similar EFS (HR = 1.04; 95% CI: 0.62-1.72). CONCLUSIONS RCB I experienced a poorer prognosis compared to RCB 0 in the general population, a pattern also observed in the HER2+ subtype and TNBC. However, no significant prognostic disparity was noted between RCB 0 and RCB I in the luminal subtype.
Collapse
Affiliation(s)
- Xinlong Tao
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University and Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China.
| | - Jingqi Han
- Department of Pathology, Affiliated Hospital of Qinghai University, Xining, 810000, China.
| | - Yongxin Li
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University and Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China.
| | - Yaming Tian
- Department of Imaging, Affiliated Hospital of Qinghai University, Xining, 810000, China.
| | - Zhou Juan Li
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University and Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China.
| | - Jinming Li
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University and Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China.
| | - Xinjian Guo
- Department of Pathology, Affiliated Hospital of Qinghai University, Xining, 810000, China.
| | - Jiuda Zhao
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University and Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China.
| |
Collapse
|
3
|
Ren K, Wang S, Ye T, Zhu Z, Hong S, Wang S, Liu J. Efficacy and safety of Anlotinib based neoadjuvant chemotherapy for locally advanced triple negative breast cancer (TNBC). BMC Cancer 2024; 24:1237. [PMID: 39375657 PMCID: PMC11459846 DOI: 10.1186/s12885-024-12852-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 08/26/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND Anlotinib, an oral multitarget tyrosine kinase inhibitor, has shown the ability to inhibit tumor angiogenesis. This study aimed to assess the effectiveness and safety of anlotinib plus docetaxel, epirubicin, and cyclophosphamide (TEC) as a neoadjuvant chemotherapy regimen for locally advanced TNBC. METHODS Locally advanced TNBC patients who had received no prior systemic treatment were eligible for this study. The enrolled patients were scheduled to undergo six cycles of anlotinib (12 mg, d1-14, q3w) plus docetaxel (75 mg/m2, d1, q3w), epirubicin (75 mg/m2, d1, q3w) and cyclophosphamide (600 mg/m2, d1, q3w) prior to surgery, unless there was disease progression or severe toxicity. The primary objective of this study was the safety of this therapeutic regimen, and the secondary objective was the tumor response. The safety of this regimen was assessed using Common Terminology Criteria for Adverse Events (CTCAE) version 4.03 and the efficacy of this treatment was measured using the Response Evaluation Criteria in Solid Tumors version 1.1. RESULTS A total of 18 patients were included in this study. Participants completed an average of 5.56 neoadjuvant treatment cycles. The objective response rate (ORR) was 83.33%, and the disease control rate was 100%, respectively. The pCR was 55.6%. No patients discontinued therapy because of Adverse effects (AEs). Grade 3 or 4 AEs were observed in 5 cases (27.8%), with neutropenia and palmar-plantar erythrodysesthesia syndrome being the most common. CONCLUSIONS Anlotinib combined with TEC as neoadjuvant therapy demonstrated manageable toxicity and promising antitumor activity for locally advanced TNBC. Further investigation of this combination regimen is warranted.
Collapse
Affiliation(s)
- Kuojun Ren
- Breast Cancer Center, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Breast Cancer Center, Anhui Provincial Cancer Hospital, Hefei, China
| | - Shuhan Wang
- Breast Cancer Center, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Breast Cancer Center, Anhui Provincial Cancer Hospital, Hefei, China
| | - Tingbo Ye
- Breast Cancer Center, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhengzhi Zhu
- Breast Cancer Center, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Breast Cancer Center, Anhui Provincial Cancer Hospital, Hefei, China
| | - Shikai Hong
- Breast Cancer Center, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Breast Cancer Center, Anhui Provincial Cancer Hospital, Hefei, China
| | - Shengying Wang
- Breast Cancer Center, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Breast Cancer Center, Anhui Provincial Cancer Hospital, Hefei, China.
| | - Jianjun Liu
- Breast Cancer Center, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Breast Cancer Center, Anhui Provincial Cancer Hospital, Hefei, China.
| |
Collapse
|
4
|
Vaklavas C, Matsen CB, Chu Z, Boucher KM, Scherer SD, Pathi S, Beck A, Brownson KE, Buys SS, Chittoria N, D'Astous E, Gulbahce HE, Henry NL, Kimani S, Porretta J, Rosenthal R, Ward J, Wei M, Welm BE, Welm AL. TOWARDS Study: Patient-Derived Xenograft Engraftment Predicts Poor Survival in Patients With Newly Diagnosed Triple-Negative Breast Cancer. JCO Precis Oncol 2024; 8:e2300724. [PMID: 39074345 PMCID: PMC11371112 DOI: 10.1200/po.23.00724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 05/04/2024] [Accepted: 05/28/2024] [Indexed: 07/31/2024] Open
Abstract
PURPOSE Assessing risk of recurrence for nonmetastatic triple-negative breast cancer (TNBC) is a key determinant of therapeutic strategy. The best predictor of recurrence risk is failure to achieve a pathologic complete response after preoperative chemotherapy, but it imperfectly correlates with the definitive end points of relapse-free and overall survival (OS). The inability to accurately predict recurrence has led to increasingly toxic treatment regimens for patients with early-stage TNBC. Better assays for recurrence risk are needed to tailor aggressive therapy for patients who need it and avoid overtreatment and unnecessary toxicity for those at low risk. The purpose of this study was to determine if patient-derived xenograft (PDX) engraftment of newly diagnosed breast tumors can serve as an accurate predictor of recurrence and death from breast cancer. METHODS This study was a blinded noninterventional trial comprising 80 patients with newly diagnosed, nonmetastatic, estrogen receptor (ER)-negative or ER-low breast cancer. RESULTS PDX engraftment was strongly associated with relapse in 1 year: 8 of 18 (44.4%) patients whose tumors engrafted relapsed versus 1 of 62 (1.6%) patients whose tumors did not engraft (P < .0001). Patients whose tumors engrafted had a hazard ratio (HR) for relapse of 17.5. HRs for OS and breast cancer-specific survival in PDX+ patients were 21.1 and 39.5, respectively. CONCLUSION We report that the ability of a tumor to engraft as a PDX predicts early recurrence by serving as a functional readout of aggressiveness and prospectively identifies the most devastating tumors. This provides new opportunity to develop surrogate assays, such as biomarkers of engraftment, which will extend the clinical feasibility of this finding.
Collapse
Affiliation(s)
- Christos Vaklavas
- Department of Internal Medicine, Division of Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Cindy B Matsen
- Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Zhengtao Chu
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Kenneth M Boucher
- Department of Internal Medicine, Division of Epidemiology, Salt Lake City, UT
| | - Sandra D Scherer
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Satya Pathi
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Anna Beck
- Department of Internal Medicine, Division of Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Kirstyn E Brownson
- Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Saundra S Buys
- Department of Internal Medicine, Division of Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Namita Chittoria
- Department of Internal Medicine, Division of Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Elyse D'Astous
- Huntsman Cancer Institute Clinical Trials Office, Salt Lake City, UT
| | - H Evin Gulbahce
- Department of Pathology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - N Lynn Henry
- Department of Internal Medicine, Division of Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
- Current Address: Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Stephen Kimani
- Department of Internal Medicine, Division of Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Jane Porretta
- Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Regina Rosenthal
- Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - John Ward
- Department of Internal Medicine, Division of Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Mei Wei
- Department of Internal Medicine, Division of Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Bryan E Welm
- Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Alana L Welm
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| |
Collapse
|
5
|
Liu Z, Li J, Zhao F, Ren D, Li Z, Chen Y, Huang S, Liu Z, Zhao Y, Wang M, Li H, Xu Z, Shen G, Zhao J. Long-term survival after neoadjuvant therapy for triple-negative breast cancer under different treatment regimens: a systematic review and network meta-analysis. BMC Cancer 2024; 24:440. [PMID: 38594636 PMCID: PMC11005293 DOI: 10.1186/s12885-024-12222-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 04/02/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is a life-threatening subtype of breast cancer with limited treatment options. Therefore, this network meta-analysis (NMA) aimed to evaluate and compare the effect of various neoadjuvant chemotherapy (NCT) options on the long-term survival of patients with TNBC. METHODS PubMed, Embase, Medline, Cochrane Library, Web of Science, and major international conference databases were systematically searched for randomized controlled trials (RCTs) on the efficacy of various NCT options in patients with TNBC. Searches were performed from January 2000 to June 2023. Study heterogeneity was assessed using the I2 statistic. Hazard ratios (HRs) and 95% confidence intervals (CIs) were used to evaluate disease-free survival (DFS) and overall survival (OS). Odds ratios (ORs) and 95% CIs were used to evaluate the pathologic complete response (pCR). The primary outcome was DFS. RESULTS We conducted an NMA of 21 RCTs involving 8873 patients with TNBC. Our study defined the combination of anthracyclines and taxanes as the preferred treatment option. On this basis, the addition of any of the following new drugs is considered a new treatment option: bevacizumab (B), platinum (P), poly-ADP-ribose polymerase inhibitors (PARPi), and immune checkpoint inhibitor (ICI). Based on the surface under the cumulative ranking curve (SUCRA) values, the top three SUCRA area values of DFS were taxanes, anthracycline, and cyclophosphamide (TAC; 89.23%); CT (84.53%); and B (81.06%). The top three SUCRA area values of OS were CT (83.70%), TAC (62.02%), and B-containing regimens (60.06%). The top three SUCRA area values of pCR were B + P-containing regimens (82.7%), ICI + P-containing regimens (80.2%), and ICI-containing regimens (61.8%). CONCLUSIONS This NMA showed that standard chemotherapy is a good choice with respect to long-term survival. Moreover, B associated with P-containing regimens is likely to be the optimal treatment option for neoadjuvant TNBC in terms of pCR.
Collapse
Affiliation(s)
- Zhilin Liu
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai University, People's Republic of China, Qinghai Provincial Clinical Research Center for Cancer, Qinghai Provincial Institute of Cancer Research, Xining, China
| | - Jinming Li
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai University, People's Republic of China, Qinghai Provincial Clinical Research Center for Cancer, Qinghai Provincial Institute of Cancer Research, Xining, China
| | - Fuxing Zhao
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai University, People's Republic of China, Qinghai Provincial Clinical Research Center for Cancer, Qinghai Provincial Institute of Cancer Research, Xining, China
| | - Dengfeng Ren
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai University, People's Republic of China, Qinghai Provincial Clinical Research Center for Cancer, Qinghai Provincial Institute of Cancer Research, Xining, China
| | - Zitao Li
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai University, People's Republic of China, Qinghai Provincial Clinical Research Center for Cancer, Qinghai Provincial Institute of Cancer Research, Xining, China
| | - Yongzhi Chen
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai University, People's Republic of China, Qinghai Provincial Clinical Research Center for Cancer, Qinghai Provincial Institute of Cancer Research, Xining, China
| | - Shifen Huang
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai University, People's Republic of China, Qinghai Provincial Clinical Research Center for Cancer, Qinghai Provincial Institute of Cancer Research, Xining, China
| | - Zhen Liu
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai University, People's Republic of China, Qinghai Provincial Clinical Research Center for Cancer, Qinghai Provincial Institute of Cancer Research, Xining, China
| | - Yi Zhao
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai University, People's Republic of China, Qinghai Provincial Clinical Research Center for Cancer, Qinghai Provincial Institute of Cancer Research, Xining, China
| | - Miaozhou Wang
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai University, People's Republic of China, Qinghai Provincial Clinical Research Center for Cancer, Qinghai Provincial Institute of Cancer Research, Xining, China
| | - Huihui Li
- Department of Breast Medical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | | | - Guoshuang Shen
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai University, People's Republic of China, Qinghai Provincial Clinical Research Center for Cancer, Qinghai Provincial Institute of Cancer Research, Xining, China.
| | - Jiuda Zhao
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai University, People's Republic of China, Qinghai Provincial Clinical Research Center for Cancer, Qinghai Provincial Institute of Cancer Research, Xining, China.
| |
Collapse
|
6
|
Pala L, Sala I, Pagan E, De Pas T, Zattarin E, Catania C, Cocorocchio E, Rossi G, Laszlo D, Ceresoli G, Canzian J, Valenzi E, Bagnardi V, Conforti F. "Heterogeneity of treatment effect on patients' long-term outcome according to pathological response type in neoadjuvant RCTs for breast cancer.". Breast 2024; 73:103672. [PMID: 38244459 PMCID: PMC10831306 DOI: 10.1016/j.breast.2024.103672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/02/2023] [Accepted: 01/08/2024] [Indexed: 01/22/2024] Open
Abstract
INTRODUCTION To provide evidence explaining the poor association between pCR and patients' long-term outcome at trial-level in neoadjuvant RCTs for breast cancer (BC), we performed a systematic-review and meta-analysis of all RCTs testing neoadjuvant treatments for early-BC and reporting the hazard ratio of DFS (HRDFS) for the intervention versus control arm stratified by pathological response type (i.e., pCR yes versus no). METHODS The objective was to explore differences of treatment effects on DFS across patients with and without pCR. We calculated the pooled HRDFS in the two strata of pathological response (i.e., pCR yes versus no) using a random-effects model, and assessed the difference between these two estimates using an interaction test. RESULTS Ten RCTs and 8496 patients were included in the analysis. Patients obtaining pCR in the intervention-arm had a higher, although not statistically significant, risk of DFS-event as compared with patients obtaining pCR in the control-arm: the pooled HRDFS for the experimental versus control arm was 1.23 (95%CI, 0.91-1.65). On the opposite, the risk of DFS-event was higher for control as compared with the intervention-arm in the stratum of patients without pCR: the pooled HRDFS was 0.86 (95%CI, 0.78-0.95). Treatment effect on DFS was significantly different according to pathological response type (interaction test p: 0.014). CONCLUSION We reported new evidence that contributes to explaining the poor surrogacy value of pCR at trial-level in neoadjuvant RCTs for early-BC.
Collapse
Affiliation(s)
- Laura Pala
- Department of Medical Oncology, Humanitas Gavazzeni, Bergamo, Italy
| | - Isabella Sala
- Department of Statistics and Quantitative Methods, University of Milan-Bicocca, Milan, Italy; Department of Medicine and Surgery, University of Milan-Bicocca, Milan, Italy
| | - Eleonora Pagan
- Department of Statistics and Quantitative Methods, University of Milan-Bicocca, Milan, Italy
| | - Tommaso De Pas
- Department of Medical Oncology, Humanitas Gavazzeni, Bergamo, Italy
| | - Emma Zattarin
- Department of Medical Oncology, Humanitas Gavazzeni, Bergamo, Italy
| | - Chiara Catania
- Department of Medical Oncology, Humanitas Gavazzeni, Bergamo, Italy
| | | | - Giovanna Rossi
- Department of Medical Oncology, Humanitas Gavazzeni, Bergamo, Italy
| | - Daniele Laszlo
- Department of Medical Oncology, Humanitas Gavazzeni, Bergamo, Italy
| | | | - Jacopo Canzian
- Department of Medical Oncology, Humanitas Gavazzeni, Bergamo, Italy
| | - Elena Valenzi
- Department of Medical Oncology, Humanitas Gavazzeni, Bergamo, Italy
| | - Vincenzo Bagnardi
- Department of Statistics and Quantitative Methods, University of Milan-Bicocca, Milan, Italy
| | - Fabio Conforti
- Department of Medical Oncology, Humanitas Gavazzeni, Bergamo, Italy; Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy.
| |
Collapse
|
7
|
Bhardwaj PV, Wang Y, Brunk E, Spanheimer PM, Abdou YG. Advances in the Management of Early-Stage Triple-Negative Breast Cancer. Int J Mol Sci 2023; 24:12478. [PMID: 37569851 PMCID: PMC10419523 DOI: 10.3390/ijms241512478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer with both inter- and intratumor heterogeneity, thought to result in a more aggressive course and worse outcomes. Neoadjuvant therapy (NAT) has become the preferred treatment modality of early-stage TNBC as it allows for the downstaging of tumors in the breast and axilla, monitoring early treatment response, and most importantly, provides important prognostic information that is essential to determining post-surgical therapies to improve outcomes. It focuses on combinations of systemic drugs to optimize pathologic complete response (pCR). Excellent response to NAT has allowed surgical de-escalation in ideal candidates. Further, treatment algorithms guide the systemic management of patients based on their pCR status following surgery. The expanding knowledge of molecular pathways, genomic sequencing, and the immunological profile of TNBC has led to the use of immune checkpoint inhibitors and targeted agents, including PARP inhibitors, further revolutionizing the therapeutic landscape of this clinical entity. However, subgroups most likely to benefit from these novel approaches in TNBC remain elusive and are being extensively studied. In this review, we describe current practices and promising therapeutic options on the horizon for TNBC, surgical advances, and future trends in molecular determinants of response to therapy in early-stage TNBC.
Collapse
Affiliation(s)
- Prarthna V. Bhardwaj
- Division of Hematology-Oncology, University of Massachusetts Chan Medical School—Baystate, Springfield, MA 01199, USA
| | - Yue Wang
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Curriculum in Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Elizabeth Brunk
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Integrative Program for Biological and Genomic Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, UNC Chapel Hill, NC 27599, USA
- Computational Medicine Program, UNC Chapel Hill, NC 27599, USA
| | - Philip M. Spanheimer
- Lineberger Comprehensive Cancer Center, UNC Chapel Hill, NC 27599, USA
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yara G. Abdou
- Lineberger Comprehensive Cancer Center, UNC Chapel Hill, NC 27599, USA
- Division of Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
8
|
Taurelli Salimbeni B, Corvaja C, Valenza C, Zagami P, Curigliano G. The triple negative breast cancer drugs graveyard: a review of failed clinical trials 2017-2022. Expert Opin Investig Drugs 2022; 31:1203-1226. [PMID: 36413823 DOI: 10.1080/13543784.2022.2151433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) accounts for 15-20% of breast cancers (BC) and has the worst prognosis. It is characterized by the absence of both hormone receptor (HR) and human epidermal growth factor receptor 2 (HER2). TNBC has more limited therapeutic options compared to other subtypes, meaning that there is still a long way to go to discover target treatments. AREAS COVERED Our review aims to summarize phase II/III clinical trials enrolling patients with TNBC that have been published between 2017 and 2022 but failed to reach their primary endpoint. We here try to emphasize the limitations and weaknesses noted in negative studies and to point out unexpected results which might be useful to enhance the therapeutic approach to TNBC disease. EXPERT OPINION A deeper understanding of the mechanisms behind TNBC heterogeneity allowed to enhance the knowledge of new prognostic and predictive biomarkers of response. However, it is also through several failed clinical trials that we were able to define new therapeutic approaches which improved TNBC patients' clinical outcomes. Nowadays, we still need to overcome several difficulties to fully recognize different intracellular and extracellular pathways that crosstalk in TNBC and the mechanisms of resistance to identify novel tailored-patients' therapies.
Collapse
Affiliation(s)
- Beatrice Taurelli Salimbeni
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, Irccs, Milan, Italy.,Department of Clinical and Molecular Medicine, Oncology Unit, "la Sapienza" University of Rome, Azienda Ospedaliera Sant'Andrea, Rome, Italy
| | - Carla Corvaja
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, Irccs, Milan, Italy.,Department of Medicine, University of Udine, Udine, Italy
| | - Carmine Valenza
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, Irccs, Milan, Italy.,Department of Oncology and Haematology, University of Milan, Milan, Italy
| | - Paola Zagami
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, Irccs, Milan, Italy.,Department of Oncology and Haematology, University of Milan, Milan, Italy.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, Irccs, Milan, Italy.,Department of Oncology and Haematology, University of Milan, Milan, Italy
| |
Collapse
|
9
|
Conforti F, Pala L, Bagnardi V, De Pas T, Colleoni M, Buyse M, Hortobagyi G, Gianni L, Winer E, Loibl S, Cortes J, Piccart M, Wolff AC, Viale G, Gelber RD. Surrogacy of Pathologic Complete Response in Trials of Neoadjuvant Therapy for Early Breast Cancer: Critical Analysis of Strengths, Weaknesses, and Misinterpretations. JAMA Oncol 2022; 8:1668-1675. [PMID: 36201176 DOI: 10.1001/jamaoncol.2022.3755] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Importance The pathologic complete response (pCR) is supported by regulatory agencies as a surrogate end point for long-term patients' clinical outcomes in the accelerated approval process of new drugs tested in neoadjuvant randomized clinical trials (RCTs) for early breast cancer (BC). However, a meaningful association between pCR and patients' survival has been proven only at the patient level (ie, significantly better survival of patients who achieved pCR compared with those who did not), but not at trial level (ie, poor association between degree of improvement in pCR rate and survival reported across trials). Observations We critically discuss the potential reasons of such discrepancy between pCR surrogacy value at the patient and trial level, as well as the relevant implications for both clinical research and drug regulatory policy. We also describe alternative surrogate end points, including combined end points that jointly analyzed pathological response and event-free survival data, or the assessment of circulating tumor DNA (ctDNA). Such proposed surrogate end points could overcome limits of pCR and provide a reasonable trade-off between the 2 conflicting needs to have access to effective therapies rapidly, and to reliably assess patients' clinical benefit. Conclusions and Relevance Using surrogate end points to grant drug approvals is justified only when they can provide accurate prediction of a drug's effect on the long-term patient outcomes. Evidence currently available does not support pCR used alone as a reliable surrogate end point in regulatory neoadjuvant RCTs for BC. The surrogacy value at trial level of potentially more robust surrogate end points needs to be urgently tested.
Collapse
Affiliation(s)
- Fabio Conforti
- European Institute of Oncology, Milan, Italy.,Department of Medical Oncology, Cliniche, Humanitas Gavazzeni, Bergamo, Italy
| | - Laura Pala
- European Institute of Oncology, Milan, Italy
| | - Vincenzo Bagnardi
- Department of Medical Oncology, Cliniche, Humanitas Gavazzeni, Bergamo, Italy.,Department of Statistics and Quantitative Methods, University of Milan-Bicocca, Milan, Italy
| | - Tommaso De Pas
- European Institute of Oncology, Milan, Italy.,Harvard T.H. Chan School of Public Health, and Frontier Science & Technology Research Foundation, Boston, Massachusetts
| | - Marco Colleoni
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Marc Buyse
- International Drug Development Institute (IDDI), Louvain-la-Neuve, Belgium
| | - Gabriel Hortobagyi
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Eric Winer
- Yale Cancer Center, New Haven, Connecticut
| | - Sibylle Loibl
- Center for Hematology and Oncology Bethanien, Frankfurt, Germany
| | - Javier Cortes
- International Breast Cancer Center, Pangaea Oncology, Quiron Group, Madrid and Barcelona, Spain.,Universidad Europea de Madrid, Faculty of Biomedical and Health Sciences, Department of Medicine, Madrid, Spain
| | - Martine Piccart
- Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | | | - Giuseppe Viale
- Department of Pathology, European Institute of Oncology, Milan, Italy.,University of Milan, Milan, Italy
| | - Richard D Gelber
- Department of Data Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.,Harvard T.H. Chan School of Public Health, and Frontier Science & Technology Research Foundation, Boston, Massachusetts
| |
Collapse
|
10
|
Shepherd JH, Ballman K, Polley MYC, Campbell JD, Fan C, Selitsky S, Fernandez-Martinez A, Parker JS, Hoadley KA, Hu Z, Li Y, Soloway MG, Spears PA, Singh B, Tolaney SM, Somlo G, Port ER, Ma C, Kuzma C, Mamounas E, Golshan M, Bellon JR, Collyar D, Hahn OM, Hudis CA, Winer EP, Partridge A, Hyslop T, Carey LA, Perou CM, Sikov WM. CALGB 40603 (Alliance): Long-Term Outcomes and Genomic Correlates of Response and Survival After Neoadjuvant Chemotherapy With or Without Carboplatin and Bevacizumab in Triple-Negative Breast Cancer. J Clin Oncol 2022; 40:1323-1334. [PMID: 35044810 PMCID: PMC9015203 DOI: 10.1200/jco.21.01506] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 11/10/2021] [Accepted: 12/13/2021] [Indexed: 12/14/2022] Open
Abstract
PURPOSE CALGB 40603 (NCT00861705), a 2 × 2 randomized phase II trial, demonstrated that adding carboplatin or bevacizumab to weekly paclitaxel (wP) followed by doxorubicin and cyclophosphamide significantly increased the pathologic complete response (pCR) rate in stage II-III triple-negative breast cancer. We now report long-term outcomes (LTOs) and correlative science end points. PATIENTS AND METHODS The Kaplan-Meier method was used to estimate LTOs in 443 patients who initiated study treatment. Log-rank tests and Cox proportional hazards models evaluated the impact of clinical characteristics, pathologic response, calculated residual cancer burden (RCB) in patients with residual disease (RD), treatment assignment, and dose delivery during wP on LTOs, including event-free survival (EFS). Genomic predictors of treatment response and outcomes were assessed on pretreatment tumor samples by mRNA sequencing. RESULTS Among baseline characteristics, only the clinical stage was associated with LTOs. At a median follow-up of 7.9 years, LTOs were not significantly improved with either carboplatin or bevacizumab, overall or in patients with basal-like subtype cancers by genomic analysis. Patients with pCR (n = 205, 46.3%) had significantly higher 5-year EFS (85.5% v 56.6%, log-rank P < .0001) and overall survival (87.9% v 63.4%, P < .0001) rates compared with patients with RD, even those with RCB class I. Among clinical and genomic features, evidence of immune activation, including tumor-infiltrating lymphocytes and low B-cell receptor evenness, was associated with pCR and improved EFS. CONCLUSION Despite higher pCR rates, neither carboplatin nor bevacizumab appeared to improve LTOs although the study was not powered to assess these secondary end points. pCR was associated with superior LTOs even when compared with minimal RD. Markers of immune activation in pretreatment tumor biopsies were independently associated with higher pCR rates and improved survival.
Collapse
Affiliation(s)
- Jonathan H. Shepherd
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Karla Ballman
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, MN
| | - Mei-Yin C. Polley
- Department of Public Health Sciences, University of Chicago, Chicago, IL
| | - Jordan D. Campbell
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, MN
| | - Cheng Fan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | | | | | | | - Katherine A. Hoadley
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Genetics, University of North Carolina, Chapel Hill, NC
| | - Zhiyuan Hu
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Yan Li
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Matthew G. Soloway
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Patricia A. Spears
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | | | | | - George Somlo
- City of Hope Comprehensive Cancer Center, Duarte, CA
| | | | - Cynthia Ma
- Washington University School of Medicine, St Louis, MO
| | - Charles Kuzma
- FirstHealth Sanford Hematology and Oncology, Sanford, NC
| | | | - Mehra Golshan
- Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | | | | | | | | | | | | | - Terry Hyslop
- Department of Biostatistics & Bioinformatics, School of Medicine, Duke University, Durham, NC
| | - Lisa A. Carey
- Division of Hematology-Oncology, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Charles M. Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Genetics, University of North Carolina, Chapel Hill, NC
| | - William M. Sikov
- Program in Women's Oncology, Women and Infants Hospital of Rhode Island and Warren Alpert Medical School of Brown University, Providence, RI
| |
Collapse
|
11
|
Yau C, Osdoit M, van der Noordaa M, Shad S, Wei J, de Croze D, Hamy AS, Laé M, Reyal F, Sonke GS, Steenbruggen TG, van Seijen M, Wesseling J, Martín M, Del Monte-Millán M, López-Tarruella S, Boughey JC, Goetz MP, Hoskin T, Gould R, Valero V, Edge SB, Abraham JE, Bartlett JMS, Caldas C, Dunn J, Earl H, Hayward L, Hiller L, Provenzano E, Sammut SJ, Thomas JS, Cameron D, Graham A, Hall P, Mackintosh L, Fan F, Godwin AK, Schwensen K, Sharma P, DeMichele AM, Cole K, Pusztai L, Kim MO, van 't Veer LJ, Esserman LJ, Symmans WF. Residual cancer burden after neoadjuvant chemotherapy and long-term survival outcomes in breast cancer: a multicentre pooled analysis of 5161 patients. Lancet Oncol 2022; 23:149-160. [PMID: 34902335 PMCID: PMC9455620 DOI: 10.1016/s1470-2045(21)00589-1] [Citation(s) in RCA: 260] [Impact Index Per Article: 86.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Previous studies have independently validated the prognostic relevance of residual cancer burden (RCB) after neoadjuvant chemotherapy. We used results from several independent cohorts in a pooled patient-level analysis to evaluate the relationship of RCB with long-term prognosis across different phenotypic subtypes of breast cancer, to assess generalisability in a broad range of practice settings. METHODS In this pooled analysis, 12 institutes and trials in Europe and the USA were identified by personal communications with site investigators. We obtained participant-level RCB results, and data on clinical and pathological stage, tumour subtype and grade, and treatment and follow-up in November, 2019, from patients (aged ≥18 years) with primary stage I-III breast cancer treated with neoadjuvant chemotherapy followed by surgery. We assessed the association between the continuous RCB score and the primary study outcome, event-free survival, using mixed-effects Cox models with the incorporation of random RCB and cohort effects to account for between-study heterogeneity, and stratification to account for differences in baseline hazard across cancer subtypes defined by hormone receptor status and HER2 status. The association was further evaluated within each breast cancer subtype in multivariable analyses incorporating random RCB and cohort effects and adjustments for age and pretreatment clinical T category, nodal status, and tumour grade. Kaplan-Meier estimates of event-free survival at 3, 5, and 10 years were computed for each RCB class within each subtype. FINDINGS We analysed participant-level data from 5161 patients treated with neoadjuvant chemotherapy between Sept 12, 1994, and Feb 11, 2019. Median age was 49 years (IQR 20-80). 1164 event-free survival events occurred during follow-up (median follow-up 56 months [IQR 0-186]). RCB score was prognostic within each breast cancer subtype, with higher RCB score significantly associated with worse event-free survival. The univariable hazard ratio (HR) associated with one unit increase in RCB ranged from 1·55 (95% CI 1·41-1·71) for hormone receptor-positive, HER2-negative patients to 2·16 (1·79-2·61) for the hormone receptor-negative, HER2-positive group (with or without HER2-targeted therapy; p<0·0001 for all subtypes). RCB score remained prognostic for event-free survival in multivariable models adjusted for age, grade, T category, and nodal status at baseline: the adjusted HR ranged from 1·52 (1·36-1·69) in the hormone receptor-positive, HER2-negative group to 2·09 (1·73-2·53) in the hormone receptor-negative, HER2-positive group (p<0·0001 for all subtypes). INTERPRETATION RCB score and class were independently prognostic in all subtypes of breast cancer, and generalisable to multiple practice settings. Although variability in hormone receptor subtype definitions and treatment across patients are likely to affect prognostic performance, the association we observed between RCB and a patient's residual risk suggests that prospective evaluation of RCB could be considered to become part of standard pathology reporting after neoadjuvant therapy. FUNDING National Cancer Institute at the US National Institutes of Health.
Collapse
Affiliation(s)
- Christina Yau
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA.
| | - Marie Osdoit
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA; Department of Surgery, Institut Curie, Paris, France
| | | | - Sonal Shad
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Jane Wei
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Diane de Croze
- Department of Tumor Biology, Institut Curie, Paris, France
| | | | - Marick Laé
- Department of Tumor Biology, Institut Curie, Paris, France; Department of Pathology, Université de Rouen Normandie, Rouen, France
| | - Fabien Reyal
- Department of Surgery, Institut Curie, Paris, France
| | - Gabe S Sonke
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Tessa G Steenbruggen
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Maartje van Seijen
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Jelle Wesseling
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Miguel Martín
- Department of Medical Oncology, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Maria Del Monte-Millán
- Department of Medical Oncology, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Sara López-Tarruella
- Department of Medical Oncology, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | | | | | - Tanya Hoskin
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Rebekah Gould
- Department of Pathology and Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vicente Valero
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stephen B Edge
- Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Jean E Abraham
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - John M S Bartlett
- Diagnostic Development Program, Ontario Institute for Cancer Research, Toronto, Canada; Deanery of Molecular, Genetic and Population Health Sciences, Edinburgh Cancer Research Centre, Edinburgh, UK; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Carlos Caldas
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - Janet Dunn
- Warwick Clinical Trials Unit, University of Warwick, Coventry, UK
| | - Helena Earl
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - Larry Hayward
- Department of Oncology, Western General Hospital, Edinburgh, UK
| | - Louise Hiller
- Warwick Clinical Trials Unit, University of Warwick, Coventry, UK
| | - Elena Provenzano
- Department of Histopathology, University of Cambridge, Cambridge, UK
| | | | - Jeremy S Thomas
- Department of Pathology, Western General Hospital, Edinburgh, UK
| | - David Cameron
- Department of Oncology, Western General Hospital, Edinburgh, UK
| | - Ashley Graham
- Department of Pathology, Western General Hospital, Edinburgh, UK
| | - Peter Hall
- Department of Oncology, Western General Hospital, Edinburgh, UK
| | - Lorna Mackintosh
- Department of Pathology, Western General Hospital, Edinburgh, UK
| | - Fang Fan
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Kelsey Schwensen
- Department of Medical Oncology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Priyanka Sharma
- Department of Medical Oncology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Angela M DeMichele
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kimberly Cole
- Department of Pathology, Yale University, New Haven, CT, USA
| | - Lajos Pusztai
- Department of Medical Oncology, Yale University, New Haven, CT, USA
| | - Mi-Ok Kim
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Laura J van 't Veer
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Laura J Esserman
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - W Fraser Symmans
- Department of Pathology and Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
12
|
Conforti F, Pala L, Sala I, Oriecuia C, De Pas T, Specchia C, Graffeo R, Pagan E, Queirolo P, Pennacchioli E, Colleoni M, Viale G, Bagnardi V, Gelber RD. Evaluation of pathological complete response as surrogate endpoint in neoadjuvant randomised clinical trials of early stage breast cancer: systematic review and meta-analysis. BMJ 2021; 375:e066381. [PMID: 34933868 PMCID: PMC8689398 DOI: 10.1136/bmj-2021-066381] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/16/2021] [Indexed: 01/12/2023]
Abstract
OBJECTIVE To evaluate pathological complete response as a surrogate endpoint for disease-free survival and overall survival in regulatory neoadjuvant trials of early stage breast cancer. DESIGN Systematic review and meta-analysis. DATA SOURCES Medline, Embase, and Scopus to 1 December 2020. ELIGIBILITY CRITERIA FOR STUDY SELECTION Randomised clinical trials that tested neoadjuvant chemotherapy given alone or combined with other treatments, including anti-human epidermal growth factor 2 (anti-HER2) drugs, targeted treatments, antivascular agents, bisphosphonates, and immune checkpoint inhibitors. DATA EXTRACTION AND SYNTHESIS Trial level associations between the surrogate endpoint pathological complete response and disease-free survival and overall survival. METHODS A weighted regression analysis was performed on log transformed treatment effect estimates (hazard ratio for disease-free survival and overall survival and relative risk for pathological complete response), and the coefficient of determination (R2) was used to quantify the association. The secondary objective was to explore heterogeneity of results in preplanned subgroups analysis, stratifying trials according treatment type in the experimental arm, definition used for pathological complete response (breast and lymph nodes v breast only), and biological features of the disease (HER2 positive or triple negative breast cancer). The surrogate threshold effect was also evaluated, indicating the minimum value of the relative risk for pathological complete response necessary to confidently predict a non-null effect on hazard ratio for disease-free survival or overall survival. RESULTS 54 randomised clinical trials comprising a total of 32 611 patients were included in the analysis. A weak association was observed between the log(relative risk) for pathological complete response and log(hazard ratio) for both disease-free survival (R2=0.14, 95% confidence interval 0.00 to 0.29) and overall survival (R2 =0.08, 0.00 to 0.22). Similar results were found across all subgroups evaluated, independently of the definition used for pathological complete response, treatment type in the experimental arm, and biological features of the disease. The surrogate threshold effect was 5.19 for disease-free survival but was not estimable for overall survival. Consistent results were confirmed in three sensitivity analyses: excluding small trials (<200 patients enrolled), excluding trials with short median follow-up (<24 months), and replacing the relative risk for pathological complete response with the absolute difference of pathological complete response rates between treatment arms. CONCLUSION A lack of surrogacy of pathological complete response was identified at trial level for both disease-free survival and overall survival. The findings suggest that pathological complete response should not be used as primary endpoint in regulatory neoadjuvant trials of early stage breast cancer.
Collapse
Affiliation(s)
- Fabio Conforti
- Division of Melanoma, Sarcomas and Rare Tumors, IEO, European Institute of Oncology, IRCCS, Milan, Italy
| | - Laura Pala
- Division of Melanoma, Sarcomas and Rare Tumors, IEO, European Institute of Oncology, IRCCS, Milan, Italy
| | - Isabella Sala
- Department of Statistics and Quantitative Methods, University of Milan-Bicocca, Milan, Italy
| | - Chiara Oriecuia
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Tommaso De Pas
- Division of Melanoma, Sarcomas and Rare Tumors, IEO, European Institute of Oncology, IRCCS, Milan, Italy
| | - Claudia Specchia
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Rossella Graffeo
- Breast Unit of Southern Switzerland, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Eleonora Pagan
- Department of Statistics and Quantitative Methods, University of Milan-Bicocca, Milan, Italy
| | - Paola Queirolo
- Division of Melanoma, Sarcomas and Rare Tumors, IEO, European Institute of Oncology, IRCCS, Milan, Italy
| | - Elisabetta Pennacchioli
- Division of Melanoma, Sarcomas and Rare Tumors, IEO, European Institute of Oncology, IRCCS, Milan, Italy
| | - Marco Colleoni
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Giuseppe Viale
- Department of Pathology, IEO, European Institute of Oncology, IRCCS, Milan, Italy
- University of Milan, Milan, Italy
| | - Vincenzo Bagnardi
- Department of Statistics and Quantitative Methods, University of Milan-Bicocca, Milan, Italy
| | - Richard D Gelber
- Medical School, Harvard T H Chan School of Public Health, and Frontier Science and Technology Research Foundation, Boston, MA, USA
| |
Collapse
|
13
|
Gion M, Pérez-García JM, Llombart-Cussac A, Sampayo-Cordero M, Cortés J, Malfettone A. Surrogate endpoints for early-stage breast cancer: a review of the state of the art, controversies, and future prospects. Ther Adv Med Oncol 2021; 13:17588359211059587. [PMID: 34868353 PMCID: PMC8640314 DOI: 10.1177/17588359211059587] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/25/2021] [Indexed: 01/07/2023] Open
Abstract
Drug approval for early-stage breast cancer (EBC) has been historically granted in the context of registration trials based on adequate outcomes such as disease-free survival and overall survival. Improvements in long-term outcomes have made it more difficult to demonstrate the clinical benefit of a new cancer drug in large, randomized, comparative clinical trials. Therefore, the use of surrogate endpoints rather than traditional measures allows for cancer drug trials to proceed with smaller sample sizes and shorter follow-up periods, which reduces drug development time. Among surrogate endpoints for breast cancer, the increase in pathological complete response (pCR) rates was considered appropriate for accelerated drug approval. The association between pCR and long-term outcomes was strongest in patients with aggressive tumor subtypes, such as triple-negative and human epidermal growth factor receptor 2 (HER2)-positive/hormone receptor-negative breast cancers. Whereas in hormone receptor-positive/HER2-negative EBC, the most accepted surrogate markers for endocrine therapy-based trials include changes in Ki67 and the preoperative endocrine prognostic index. Beyond the classic endpoints, further prognostic tools are required to provide EBC patients with individualized and effective therapies, and the neoadjuvant setting provides an excellent platform for drug development and biomarker discovery. Nowadays, the availability of multigene signatures is offering a standardized quantitative and reproducible tool to potentiate the efficacy of standard treatment for high-risk patients and develop de-escalated treatments for patients at lower risk of relapse. In this article, we first evaluate the surrogacies used for long-term outcomes and the underlying evidence supporting the use of each surrogate endpoint for the accelerated or regular drug approval process in EBC. Next, we provide an overview of the most recent studies and innovative strategies in a (neo)adjuvant setting as a platform to accelerate new drug approval. Finally, we highlight some clinical trials aimed at tailoring systemic treatment of EBC using prognosis-related factors or early biomarkers of drug sensitivity or resistance.
Collapse
Affiliation(s)
- María Gion
- University Hospital Ramon y Cajal, Madrid, Spain
| | - José Manuel Pérez-García
- International Breast Cancer Center (IBCC), Quironsalud Group, Barcelona, Spain
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain
- Medica Scientia Innovation Research (MEDSIR), Ridgewood, NJ, USA
| | - Antonio Llombart-Cussac
- Hospital Arnau de Vilanova, Valencia, Spain
- Universidad Catolica de Valencia San Vicente Martir, Valencia, Spain
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain
- Medica Scientia Innovation Research (MEDSIR), Ridgewood, NJ, USA
| | - Miguel Sampayo-Cordero
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain
- Medica Scientia Innovation Research (MEDSIR), Ridgewood, NJ, USA
| | - Javier Cortés
- International Breast Cancer Center (IBCC), Quironsalud Group, Carrer de Vilana, 12, 08022 Barcelona, SpainVall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain
- Medica Scientia Innovation Research (MEDSIR), Ridgewood, NJ, USA
- Department of Medicine, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Madrid, Spain
| | - Andrea Malfettone
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain
- Medica Scientia Innovation Research (MEDSIR), Ridgewood, NJ, USA
| |
Collapse
|
14
|
Symmans WF, Yau C, Chen YY, Balassanian R, Klein ME, Pusztai L, Nanda R, Parker BA, Datnow B, Krings G, Wei S, Feldman MD, Duan X, Chen B, Sattar H, Khazai L, Zeck JC, Sams S, Mhawech-Fauceglia P, Rendi M, Sahoo S, Ocal IT, Fan F, LeBeau LG, Vinh T, Troxell ML, Chien AJ, Wallace AM, Forero-Torres A, Ellis E, Albain KS, Murthy RK, Boughey JC, Liu MC, Haley BB, Elias AD, Clark AS, Kemmer K, Isaacs C, Lang JE, Han HS, Edmiston K, Viscusi RK, Northfelt DW, Khan QJ, Leyland-Jones B, Venters SJ, Shad S, Matthews JB, Asare SM, Buxton M, Asare AL, Rugo HS, Schwab RB, Helsten T, Hylton NM, van 't Veer L, Perlmutter J, DeMichele AM, Yee D, Berry DA, Esserman LJ. Assessment of Residual Cancer Burden and Event-Free Survival in Neoadjuvant Treatment for High-risk Breast Cancer: An Analysis of Data From the I-SPY2 Randomized Clinical Trial. JAMA Oncol 2021; 7:1654-1663. [PMID: 34529000 DOI: 10.1001/jamaoncol.2021.3690] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Importance Residual cancer burden (RCB) distributions may improve the interpretation of efficacy in neoadjuvant breast cancer trials. Objective To compare RCB distributions between randomized control and investigational treatments within subtypes of breast cancer and explore the relationship with survival. Design, Setting, and Participants The I-SPY2 is a multicenter, platform adaptive, randomized clinical trial in the US that compares, by subtype, investigational agents in combination with chemotherapy vs chemotherapy alone in adult women with stage 2/3 breast cancer at high risk of early recurrence. Investigational treatments graduated in a prespecified subtype if there was 85% or greater predicted probability of higher rate of pathologic complete response (pCR) in a confirmatory, 300-patient, 1:1 randomized, neoadjuvant trial in that subtype. Evaluation of a secondary end point was reported from the 10 investigational agents tested in the I-SPY2 trial from March 200 through 2016, and analyzed as of September 9, 2020. The analysis plan included modeling of RCB within subtypes defined by hormone receptor (HR) and ERBB2 status and compared control treatments with investigational treatments that graduated and those that did not graduate. Interventions Neoadjuvant paclitaxel plus/minus 1 of several investigational agents for 12 weeks, then 12 weeks of cyclophosphamide/doxorubicin chemotherapy followed by surgery. Main Outcomes and Measures Residual cancer burden (pathological measure of residual disease) and event-free survival (EFS). Results A total of 938 women (mean [SD] age, 49 [11] years; 66 [7%] Asian, 103 [11%] Black, and 750 [80%] White individuals) from the first 10 investigational agents were included, with a median follow-up of 52 months (IQR, 29 months). Event-free survival worsened significantly per unit of RCB in every subtype of breast cancer (HR-positive/ERBB2-negative: hazard ratio [HZR], 1.75; 95% CI, 1.45-2.16; HR-positive/ERBB2-positive: HZR, 1.55; 95% CI, 1.18-2.05; HR-negative/ERBB2-positive: HZR, 2.39; 95% CI, 1.64-3.49; HR-negative/ERBB2-negative: HZR, 1.99; 95% CI, 1.71-2.31). Prognostic information from RCB was similar from treatments that graduated (HZR, 2.00; 95% CI, 1.57-2.55; 254 [27%]), did not graduate (HZR, 1.87; 95% CI, 1.61-2.17; 486 [52%]), or were control (HZR, 1.79; 95% CI, 1.42-2.26; 198 [21%]). Investigational treatments significantly lowered RCB in HR-negative/ERBB2-negative (graduated and nongraduated treatments) and ERBB2-positive subtypes (graduated treatments), with improved EFS (HZR, 0.61; 95% CI, 0.41-0.93) in the exploratory analysis. Conclusions and Relevance In this randomized clinical trial, the prognostic significance of RCB was consistent regardless of subtype and treatment. Effective neoadjuvant treatments shifted the distribution of RCB in addition to increasing pCR rate and appeared to improve EFS. Using a standardized quantitative method to measure response advances the interpretation of efficacy. Trial Registration ClinicalTrials.gov Identifier: NCT01042379.
Collapse
Affiliation(s)
- W Fraser Symmans
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston
| | - Christina Yau
- Department of Surgery, University of California, San Francisco
| | - Yunn-Yi Chen
- Department of Pathology, University of California, San Francisco
| | - Ron Balassanian
- Department of Pathology, University of California, San Francisco
| | - Molly E Klein
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis
| | - Lajos Pusztai
- Department of Medicine, Medical Oncology, Yale University, New Haven, Connecticut
| | - Rita Nanda
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Barbara A Parker
- Division of Hematology-Oncology, Department of Medicine, University of California, San Diego, La Jolla
| | - Brian Datnow
- Department of Pathology, University of California, San Diego, La Jolla
| | - Gregor Krings
- Department of Pathology, University of California, San Francisco
| | - Shi Wei
- Department of Anatomic Pathology, University of Alabama at Birmingham
| | - Michael D Feldman
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia
| | - Xiuzhen Duan
- Department of Pathology, Loyola University, Chicago, Illinois
| | - Beiyun Chen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Husain Sattar
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Laila Khazai
- Department of Pathology, Moffitt Cancer Center, Tampa, Florida
| | - Jay C Zeck
- Department of Pathology, Georgetown University, Washington, DC
| | - Sharon Sams
- Department of Pathology, University of Colorado Anschutz Medical Center, Aurora
| | | | - Mara Rendi
- Department of Anatomic Pathology, University of Washington, Seattle
| | - Sunati Sahoo
- Department of Pathology, University of Texas Southwestern, Dallas
| | - Idris Tolgay Ocal
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, Arizona
| | - Fang Fan
- Department of Pathology, University of Kansas Medical Center, Kansas City
| | | | - Tuyethoa Vinh
- Department of Pathology, Inova Health System, Fairfax, Virginia
| | - Megan L Troxell
- Department of Pathology, Oregon Health and Science University, Portland
| | - A Jo Chien
- Division of Hematology-Oncology, Department of Medicine, University of California, San Francisco
| | - Anne M Wallace
- Department of Surgery, University of California, San Diego, La Jolla
| | - Andres Forero-Torres
- Division of Hematology-Oncology, Department of Medicine, University of Alabama at Birmingham
| | - Erin Ellis
- Medical Oncology, Swedish Cancer Institute, Seattle, Washington
| | - Kathy S Albain
- Division of Hematology-Oncology, Department of Medicine, Loyola University Chicago Stritch School of Medicine, Chicago, Illinois
| | - Rashmi K Murthy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Judy C Boughey
- Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | - Minetta C Liu
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Barbara B Haley
- Division of Hematology-Oncology, Department of Medicine, UT Southwestern Medical Center, Dallas, Texas
| | - Anthony D Elias
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora
| | - Amy S Clark
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia
| | - Kathleen Kemmer
- Division of Hematology-Oncology, Department of Medicine, Oregon Health & Science University, Portland
| | - Claudine Isaacs
- Division of Hematology-Oncology, Department of Medicine, Georgetown University, Washington, DC
| | - Julie E Lang
- Department of Surgery, University of Southern California, Los Angeles
| | - Hyo S Han
- Department of Breast Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Kirsten Edmiston
- Department of Surgery, Inova Schar Cancer Institute, Fairfax, Virginia
| | - Rebecca K Viscusi
- Department of Surgery, University of Arizona Health Sciences, Tucson, Arizona
| | - Donald W Northfelt
- Department of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Qamar J Khan
- Division of Oncology, Department of Medicine, University of Kansas, Kansas City
| | | | - Sara J Venters
- Department of Laboratory Medicine, University of California, San Francisco
| | - Sonal Shad
- Department of Surgery, University of California, San Francisco
| | | | - Smita M Asare
- Quantum Leap Healthcare Collaborative, San Francisco, California
| | | | - Adam L Asare
- Quantum Leap Healthcare Collaborative, San Francisco, California
| | - Hope S Rugo
- Division of Hematology-Oncology, Department of Medicine, University of California, San Francisco
| | - Richard B Schwab
- Division of Hematology-Oncology, Department of Medicine, University of California, San Diego, La Jolla
| | - Teresa Helsten
- Division of Hematology-Oncology, Department of Medicine, University of California, San Diego, La Jolla
| | - Nola M Hylton
- Department of Radiology and Biomedical Imaging, University of California, San Francisco
| | - Laura van 't Veer
- Department of Laboratory Medicine, University of California, San Francisco
| | | | - Angela M DeMichele
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia
| | - Douglas Yee
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis
| | | | | |
Collapse
|
15
|
Ge J, Zuo W, Chen Y, Shao Z, Yu K. The advance of adjuvant treatment for triple-negative breast cancer. Cancer Biol Med 2021; 19:j.issn.2095-3941.2020.0752. [PMID: 34448553 PMCID: PMC8832962 DOI: 10.20892/j.issn.2095-3941.2020.0752] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 04/28/2021] [Indexed: 11/11/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer characterized by its highly aggressive behavior, early recurrence, and poor outcomes, when compared with other subtypes. Due to the absence of the estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 expression, TNBC lacks meaningful biomarkers and an effective therapeutic strategy. Chemotherapy remains the main adjuvant treatment for patients with TNBC. Anthracycline/taxane-based regimens are the standard of care in adjuvant settings. The addition of capecitabine or platinum may offer extra benefits to patients with TNBC, but at the cost of increased toxicity or adverse events. Dose-dense chemotherapy may enhance treatment efficacy in patients who are able to tolerate the treatment regimen, especially in high-risk patients. As a heterogenous disease, TNBC can be classified into several molecular subtypes according to genomic or transcriptional features, which may indicate potential targets for more precise and individualized treatment strategies. With our increased understanding of signal pathways associated with TNBC, as well as the discovery of novel biomarkers indicative of TNBC prognosis, several new therapeutic options are under investigation, and some have already reported good results. In this review, we summarized the current conventional therapeutic strategies and emerging clinical trials regarding adjuvant treatment for TNBC. Furthermore, we evaluated the prognostic value of several potential targets and the progress of targeted therapy in TNBC, both in neoadjuvant and adjuvant settings.
Collapse
Affiliation(s)
- Jingyu Ge
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Wenjia Zuo
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yiyu Chen
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhiming Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Keda Yu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
16
|
Zhang M, Liu J, Liu G, Xing Z, Jia Z, Li J, Wang W, Wang J, Qin L, Wang X, Wang X. Anti-vascular endothelial growth factor therapy in breast cancer: Molecular pathway, potential targets, and current treatment strategies. Cancer Lett 2021; 520:422-433. [PMID: 34389434 DOI: 10.1016/j.canlet.2021.08.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/30/2021] [Accepted: 08/03/2021] [Indexed: 11/15/2022]
Abstract
As the highest incidence of female malignancy, breast cancer is likewise the leading cause of cancer-related deaths. The development of cancer relies on neo-vascularization, which provides sufficient nutrition and oxygen, and supplies a pathway for distant metastasis. Angiogenesis represents the formation of new blood vessels, and is a principal pathogenetic action in breast cancer. Vascular endothelial growth factor (VEGF) is a major angiogenesis regulator that modulates the maintenance and function of mature vascular networks. Therefore, the VEGF pathway is a promising oncotherapeutic target. This review elaborates an update on the prognostic value of VEGF in breast cancer, summarizes clinical experience and lessons of anti-VEGF therapeutics, meanwhile, provides an overview of biomarkers that predict the effectiveness of anti-angiogenic treatment.
Collapse
Affiliation(s)
- Menglu Zhang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jiaqi Liu
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Gang Liu
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zeyu Xing
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ziqi Jia
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jiaxin Li
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wenyan Wang
- Department of Breast Surgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Jie Wang
- Department of Ultrasound, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ling Qin
- Department of Breast Surgical Oncology, Cancer Hospital of HuanXing, Beijing, 100021, China
| | - Xin Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Xiang Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
17
|
Efficacy and safety of neoadjuvant immune checkpoint inhibitors in early-stage triple-negative breast cancer: a systematic review and meta-analysis. J Cancer Res Clin Oncol 2021; 147:3369-3379. [PMID: 33745080 DOI: 10.1007/s00432-021-03591-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 03/07/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE There is uncertainty regarding the role of adding immune checkpoint inhibitors (ICIs) to neoadjuvant chemotherapy (NACT) in early-stage triple-negative breast cancer (TNBC). METHODS We identified randomized controlled trials (RCTs) comparing ICIs combined with NACT to NACT in early-stage TNBC. Efficacy outcomes included pathological complete response (pCR) and event-free survival (EFS). Toxicity data included any grade 3/4 adverse events (AEs), serious AEs, AEs leading to death, common and meaningful AEs associated with chemotherapy and immune-related AEs. Odds ratio (ORs), hazard ratios (HR) and their respective 95% confidence intervals (CI) for efficacy and toxicity were extracted and pooled in a meta-analysis. Differences in the odds for pCR between programmed death ligand 1 (PD-L1) status and between PD-L1 and PD-1 inhibitors were also assessed. RESULTS Five RCTs comprising 2,075 patients were analyzed. Compared to NACT alone, combination of ICIs and NACT significantly improved pCR (OR 1.75, 95% CI 1.25-2.47, p = 0.001) and EFS (HR 0.66, 95% CI 0.48-0.91, p = 0.01). Magnitude of effect on pCR was similar between PD-L1-positive and PD-L1-negative tumors (p for the subgroup difference = 0.80) and between PD-L1 and PD-1 inhibitors (p = 0.27). The combination treatment resulted in higher odds of any grade 3/4 AEs (OR 1.31, p = 0.02) and serious AEs (OR 1.84, p = 0.006), with no statistically significant difference in AEs leading to death (OR 1.67, p = 0.51). Higher magnitude of toxicity was observed for immune-related AEs. CONCLUSION Combination of ICIs and NACT were associated with improved outcome in early-stage TNBC while increasing toxicity significantly. Longer follow-up is desired to better understand the risk and benefit ratio of this combination.
Collapse
|
18
|
Chen X, Gao Y, Zhang G, Li B, Ma T, Ma Y, Wang X. Bevacizumab Plays a double-edged role in Neoadjuvant Therapy for Non-metastatic Breast Cancer: A Systemic Review and Meta-Analysis. J Cancer 2021; 12:2643-2653. [PMID: 33854624 PMCID: PMC8040714 DOI: 10.7150/jca.53303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 02/04/2021] [Indexed: 12/04/2022] Open
Abstract
The anti-angiogenic drug Bevacizumab (Bev) is engaged in neoadjuvant therapy for non-metastatic breast cancer (NMBC). However, whether neoadjuvant Bev providing a greater benefit to patients is debatable. Our study aimed to review Bev's role in Neoadjuvant therapy (NAT) in NMBC and identify predictive markers associated with its efficacy by systemic review and meta-analysis. Eligible trials were retrieved from the Pubmed, Embase, and Cochrane Library, and random or fixed effects models were applied to synthesize data. Power of pCR to predict DFS or OS was evaluated by nonlinear mixed effect model. In NMBC, Bev significantly improved the rate of patients achieving pCR, but this benefit discontinued in DFS or OS. Biomarkers such as PAM50 intrinsic subtype, VEGF overexpression, regulation of VEGF signaling pathway, hypoxia-related genes, BRCA1/2 mutation, P53 mutation and immune phenotype can be used to predict Bev-inducing pCR and/or DFS/OS. Unfortunately, although patients with pCR survived longer than those without pCR when ignoring the use of Bev, but patients achieving pCR with Bev might survive shorter than those achieving pCR without Bev. Subgroup analyses found Bev prolonged patients' OS when given pre- and post-surgery. Lastly, adding Bev increased adverse effects. Overall, Bev offered limited effect for patients with NMBC in an unscreened population. However, in biomarkers - identified subgroup, Bev could be promising to ameliorate the prognosis of specific patients with NMBC.
Collapse
Affiliation(s)
- XinJie Chen
- Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing, 100029, China.,Oncology Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No.23 Back Road of Art Gallery, Dongcheng District, Beijing, 100010, China
| | - Yu Gao
- Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing, 100029, China.,Oncology Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No.23 Back Road of Art Gallery, Dongcheng District, Beijing, 100010, China
| | - GanLin Zhang
- Oncology Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No.23 Back Road of Art Gallery, Dongcheng District, Beijing, 100010, China
| | - BingXue Li
- Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing, 100029, China.,Oncology Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No.23 Back Road of Art Gallery, Dongcheng District, Beijing, 100010, China
| | - TingTing Ma
- Oncology Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No.23 Back Road of Art Gallery, Dongcheng District, Beijing, 100010, China
| | - YunFei Ma
- Oncology Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No.23 Back Road of Art Gallery, Dongcheng District, Beijing, 100010, China
| | - XiaoMin Wang
- Oncology Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, No.23 Back Road of Art Gallery, Dongcheng District, Beijing, 100010, China
| |
Collapse
|
19
|
Torrisi R, Marrazzo E, Agostinetto E, De Sanctis R, Losurdo A, Masci G, Tinterri C, Santoro A. Neoadjuvant chemotherapy in hormone receptor-positive/HER2-negative early breast cancer: When, why and what? Crit Rev Oncol Hematol 2021; 160:103280. [PMID: 33667658 DOI: 10.1016/j.critrevonc.2021.103280] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 02/17/2021] [Accepted: 02/27/2021] [Indexed: 12/13/2022] Open
Abstract
Indication for neoadjuvant chemotherapy (NACT) in HR+/HER2-negative tumors is controversial. Pathological complete response (pCR) rates range from 0 to 18 % while breast-conserving surgery (BCS) is achievable in up to 60 % of tumors. No pathological feature definitely predicts pCR; lobular and molecular luminal A tumors are less likely to achieve pCR although experiencing better outcomes. Luminal B subtype, high proliferation, lack of progesterone receptor, high tumor-infiltrating lymphocytes are positively associated with increased pCR rates but worse outcomes and the prognostic role of pCR is inconsistent across studies. Molecular intrinsic subtyping and genomic signatures appear as more accurate predictors of benefit from NACT, but larger studies are needed. Anthracycline and taxane-based chemotherapy remains the standard NACT; however, CDK 4/6 inhibitors and immune checkpoint inhibitors are under evaluation. In conclusion, NACT may be proposed for luminal tumors requiring downsizing for BCS after multidisciplinary evaluation, provided that other contraindications to BCS are excluded.
Collapse
Affiliation(s)
- Rosalba Torrisi
- IRCCS Humanitas Research Hospital, Dept of Medical Oncology and Hematology Unit, via Manzoni 56, Rozzano, Milan, 20089, Italy.
| | - Emilia Marrazzo
- IRCCS Humanitas Research Hospital, Breast Unit, via Manzoni 56, Rozzano, Milan, 20089, Italy
| | - Elisa Agostinetto
- IRCCS Humanitas Research Hospital, Dept of Medical Oncology and Hematology Unit, via Manzoni 56, Rozzano, Milan, 20089, Italy; Humanitas University, Department of Biomedical Sciences, Via Rita Levi Montalcini 4, Pieve Emanuele, Milan, 20090, Italy
| | - Rita De Sanctis
- IRCCS Humanitas Research Hospital, Dept of Medical Oncology and Hematology Unit, via Manzoni 56, Rozzano, Milan, 20089, Italy; Humanitas University, Department of Biomedical Sciences, Via Rita Levi Montalcini 4, Pieve Emanuele, Milan, 20090, Italy
| | - Agnese Losurdo
- IRCCS Humanitas Research Hospital, Dept of Medical Oncology and Hematology Unit, via Manzoni 56, Rozzano, Milan, 20089, Italy
| | - Giovanna Masci
- IRCCS Humanitas Research Hospital, Dept of Medical Oncology and Hematology Unit, via Manzoni 56, Rozzano, Milan, 20089, Italy
| | - Corrado Tinterri
- IRCCS Humanitas Research Hospital, Breast Unit, via Manzoni 56, Rozzano, Milan, 20089, Italy
| | - Armando Santoro
- IRCCS Humanitas Research Hospital, Dept of Medical Oncology and Hematology Unit, via Manzoni 56, Rozzano, Milan, 20089, Italy; Humanitas University, Department of Biomedical Sciences, Via Rita Levi Montalcini 4, Pieve Emanuele, Milan, 20090, Italy
| |
Collapse
|
20
|
Quintela-Fandino M, Holgado E, Manso L, Morales S, Bermejo B, Colomer R, Apala JV, Blanco R, Muñoz M, Caleiras E, Iranzo V, Martinez M, Dominguez O, Hornedo J, Gonzalez-Cortijo L, Cortes J, Gasol Cudos A, Malon D, Lopez-Alonso A, Moreno-Ortíz MC, Mouron S, Mañes S. Immuno-priming durvalumab with bevacizumab in HER2-negative advanced breast cancer: a pilot clinical trial. Breast Cancer Res 2020; 22:124. [PMID: 33176887 PMCID: PMC7661209 DOI: 10.1186/s13058-020-01362-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 10/25/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Preclinical research suggests that the efficacy of immune checkpoint inhibitors in breast cancer can be enhanced by combining them with antiangiogenics, particularly in a sequential fashion. We sought to explore the efficacy and biomarkers of combining the anti-PD-L1 durvalumab plus the antiangiogenic bevacizumab after bevacizumab monotherapy for advanced HER2-negative breast cancer. METHODS Patients had advanced HER2-negative disease that progressed while receiving single-agent bevacizumab maintenance as a part of a previous chemotherapy plus bevacizumab regimen. Treatment consisted of bi-weekly durvalumab plus bevacizumab (10 mg/kg each i.v.). Peripheral-blood mononuclear cells (PBMCs) were obtained before the first durvalumab dose and every 4 weeks and immunophenotyped by flow-cytometry. A fresh pre-durvalumab tumor biopsy was obtained; gene-expression studies and immunohistochemical staining to assess vascular normalization and characterize the immune infiltrate were conducted. Patients were classified as "non-progressors" if they had clinical benefit (SD/PR/CR) at 4 months. The co-primary endpoints were the changes in the percentage T cell subpopulations in PBMCs in progressors versus non-progressors, and PFS/OS time. RESULTS Twenty-six patients were accrued. Median PFS and OS were 3.5 and 11 months; a trend for a longer OS was detected for the hormone-positive subset (19.8 versus 7.4 months in triple-negatives; P = 0.11). Clinical benefit rate at 2 and 4 months was 60% and 44%, respectively, without significant differences between hormone-positive and triple-negative (P = 0.73). Non-progressors' tumors displayed vascular normalization features as a result of previous bevacizumab, compared with generally abnormal patterns observed in progressors. Non-progressors also showed increased T-effector and T-memory signatures and decreased TREG signatures in gene expression studies in baseline-post-bevacizumab-tumors compared with progressors. Notably, analysis of PBMC populations before durvalumab treatment was concordant with the findings in tumor samples and showed a decreased percentage of circulating TREGs in non-progressors. CONCLUSIONS This study reporting on sequential bevacizumab+durvalumab in breast cancer showed encouraging activity in a heavily pre-treated cohort. The correlative studies agree with the preclinical rationale supporting an immunopriming effect exerted by antiangiogenic treatment, probably by reducing TREGs cells both systemically and in tumor tissue. The magnitude of this benefit should be addressed in a randomized setting. TRIAL REGISTRATION (www.clinicaltrials.gov): NCT02802098 . Registered on June 16, 2020.
Collapse
MESH Headings
- Adult
- Aged
- Angiogenesis Inhibitors/administration & dosage
- Angiogenesis Inhibitors/adverse effects
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/adverse effects
- Antineoplastic Agents, Immunological/administration & dosage
- Antineoplastic Agents, Immunological/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- B7-H1 Antigen/antagonists & inhibitors
- B7-H1 Antigen/immunology
- B7-H1 Antigen/metabolism
- Bevacizumab/administration & dosage
- Bevacizumab/adverse effects
- Breast/pathology
- Breast Neoplasms/blood
- Breast Neoplasms/drug therapy
- Breast Neoplasms/immunology
- Breast Neoplasms/pathology
- Disease Progression
- Female
- Humans
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Middle Aged
- Pilot Projects
- Progression-Free Survival
- Proof of Concept Study
- Receptor, ErbB-2/analysis
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Miguel Quintela-Fandino
- Breast Cancer Clinical Research Unit - Clinical Research Program, CNIO - Spanish National Cancer Research Center, Melchor Fernandez Almagro, 3, 28029, Madrid, Spain.
- Medical Oncology Department, Hospital Universitario de Fuenlabrada, Fuenlabrada, Spain.
- Medical Oncology Department, Hospital Universitario Quiron, Pozuelo de Alarcon, Spain.
| | - Esther Holgado
- Medical Oncology Department, Hospital Universitario Ramon y Cajal, Madrid, Spain
| | - Luis Manso
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Serafin Morales
- Medical Oncology Department, Hospital Universitari Arnau Vilanova, Lleida, Spain
| | - Begoña Bermejo
- Medical Oncology Department, Hospital Clínico Universitario, Valencia, Spain
- INCLIVA, Valencia, Spain
- CIBERONC, Instituto Carlos III, Madrid, Spain
| | - Ramon Colomer
- Breast Cancer Clinical Research Unit - Clinical Research Program, CNIO - Spanish National Cancer Research Center, Melchor Fernandez Almagro, 3, 28029, Madrid, Spain
- Medical Oncology Department, Hospital Universitario La Princesa, Madrid, Spain
- Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Juan V Apala
- Breast Cancer Clinical Research Unit - Clinical Research Program, CNIO - Spanish National Cancer Research Center, Melchor Fernandez Almagro, 3, 28029, Madrid, Spain
- Medical Oncology Department, Hospital Universitario de Fuenlabrada, Fuenlabrada, Spain
| | - Raquel Blanco
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, 28049, Madrid, Spain
| | - Manuel Muñoz
- Breast Cancer Clinical Research Unit - Clinical Research Program, CNIO - Spanish National Cancer Research Center, Melchor Fernandez Almagro, 3, 28029, Madrid, Spain
| | - Eduardo Caleiras
- Histopathology Core Unit - Biotechnology Program, CNIO - Spanish National Cancer Research Center, Madrid, Spain
| | - Vega Iranzo
- CIBERONC, Instituto Carlos III, Madrid, Spain
- Medical Oncology Department, Hospital General Universitario de Valencia, Valencia, Spain
- Medicine Department, Universitat de Valencia, Valencia, Spain
| | - Mario Martinez
- Pathology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Orlando Dominguez
- Genomics Core Unit - Biotechnology Program, CNIO - Spanish National Cancer Research Center, Madrid, Spain
| | - Javier Hornedo
- Medical Oncology Department, Hospital Universitario Quiron, Pozuelo de Alarcon, Spain
| | | | - Javier Cortes
- ION Institute of Oncology, Quironsalud Group - Madrid & Barcelona, Barcelona, Spain
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Ariadna Gasol Cudos
- Medical Oncology Department, Hospital Universitari Arnau Vilanova, Lleida, Spain
| | - Diego Malon
- Medical Oncology Department, Hospital Universitario de Fuenlabrada, Fuenlabrada, Spain
| | - Antonio Lopez-Alonso
- Breast Cancer Clinical Research Unit - Clinical Research Program, CNIO - Spanish National Cancer Research Center, Melchor Fernandez Almagro, 3, 28029, Madrid, Spain
| | - María C Moreno-Ortíz
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, 28049, Madrid, Spain
| | - Silvana Mouron
- Breast Cancer Clinical Research Unit - Clinical Research Program, CNIO - Spanish National Cancer Research Center, Melchor Fernandez Almagro, 3, 28029, Madrid, Spain
| | - Santos Mañes
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, 3, 28049, Madrid, Spain.
| |
Collapse
|
21
|
Coudert B, Pierga JY, Mouret-Reynier MA, Kerrou K, Ferrero JM, Petit T, Du FL, Dupré PF, Bachelot T, Gabelle P, Chauvet MP, Coeffic D, Barbe C, Prevost JB, Paintaud G, Thibault G, Ferhat A, Dupin J, Berriolo-Riedinger A, Arnould L. Long-term outcomes in patients with PET-predicted poor-responsive HER2-positive breast cancer treated with neoadjuvant bevacizumab added to trastuzumab and docetaxel: 5-year follow-up of the randomised Avataxher study. EClinicalMedicine 2020; 28:100566. [PMID: 33205032 PMCID: PMC7649610 DOI: 10.1016/j.eclinm.2020.100566] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 09/10/2020] [Accepted: 09/11/2020] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND The open-label, randomised Phase 2 AVATAXHER study (NCT01142778) demonstrated that early PET assessment identified HER2-positive breast cancer patients who responded poorly to neoadjuvant docetaxel plus trastuzumab. Adding neoadjuvant bevacizumab for PET-predicted poor-responders improved pathological complete response (pCR) rates (43.8% vs 24.0%). We investigated long-term study outcomes. METHODS Patients were treated in three groups. All patients initially received two cycles of standard neoadjuvant therapy with [¹⁸F]-FDG PET conducted before each cycle. Those with ≥70% change in the maximum standardised uptake value (∆SUVmax) received four further cycles of standard neoadjuvant therapy (PET responders). PET-predicted poor-responders (∆SUVmax <70%) were randomised (2:1) to neoadjuvant therapy with (Group A) or without (Group B) bevacizumab for cycles 3-6. All patients received one further cycle of trastuzumab before surgery plus adjuvant trastuzumab (11 cycles). FINDINGS 142 patients were randomized and treated (PET responders, n = 69; Group A, n = 48; Group B, n = 25). 5-year disease-free survival rates were 90.5% (95% CI: 80.0-95.6%) in PET responders, 90.2% (95% CI: 75.9-96.2%) in Group A, and 76.0% (95% CI: 54.2-88.4%) in Group B. However, no difference was observed between randomised arms in a sensitivity analysis. During adjuvant therapy, the incidence of Grade ≥3 (Group A: 25.6%; Group B 12.5%) and serious adverse events (Group A: 18.6%; Group B 12.5%) was higher in Group A vs Group B, but with no apparent effect on cardiac events. INTERPRETATION In patients with HER2-positive breast cancer, an intervention based on early PET assessment and improvement of pCR does not modify disease-free survival. FUNDING Roche France.
Collapse
Affiliation(s)
- Bruno Coudert
- Centre Georges-Francois Leclerc, Dijon, France
- Corresponding author.
| | | | | | | | | | | | | | | | | | | | | | - David Coeffic
- Polyclinique Courlancy, Institut du Cancer Courlancy Reims, France
| | | | | | | | | | | | - Julien Dupin
- Roche France S.A.S., Boulogne Billancourt, France
| | | | | |
Collapse
|
22
|
Abstract
Brain metastases (BM) are the most common intracranial neoplasm and represent a major clinical challenge across many medical disciplines. The incidence of BM is increasing, largely due to improvements in primary disease therapeutics conferring greater systemic control, and advancements in neuroimaging techniques and availability leading to earlier diagnosis. In recent years, the landscape of BM treatment has changed significantly with the advent of personalized targeted chemotherapies and immunotherapy, the adoption of focal radiotherapy (RT) for higher intracranial disease burden, and the implementation of new surgical strategies. The increasing permutations of options available for the treatment of patients diagnosed with BM necessitate coordinated care by a multidisciplinary team. This review discusses the current treatment regimens for BM as well as examines the salient features of a modern multidisciplinary approach.
Collapse
|
23
|
Yee D, DeMichele AM, Yau C, Isaacs C, Symmans WF, Albain KS, Chen YY, Krings G, Wei S, Harada S, Datnow B, Fadare O, Klein M, Pambuccian S, Chen B, Adamson K, Sams S, Mhawech-Fauceglia P, Magliocco A, Feldman M, Rendi M, Sattar H, Zeck J, Ocal IT, Tawfik O, LeBeau LG, Sahoo S, Vinh T, Chien AJ, Forero-Torres A, Stringer-Reasor E, Wallace AM, Pusztai L, Boughey JC, Ellis ED, Elias AD, Lu J, Lang JE, Han HS, Clark AS, Nanda R, Northfelt DW, Khan QJ, Viscusi RK, Euhus DM, Edmiston KK, Chui SY, Kemmer K, Park JW, Liu MC, Olopade O, Leyland-Jones B, Tripathy D, Moulder SL, Rugo HS, Schwab R, Lo S, Helsten T, Beckwith H, Haugen P, Hylton NM, Van't Veer LJ, Perlmutter J, Melisko ME, Wilson A, Peterson G, Asare AL, Buxton MB, Paoloni M, Clennell JL, Hirst GL, Singhrao R, Steeg K, Matthews JB, Asare SM, Sanil A, Berry SM, Esserman LJ, Berry DA. Association of Event-Free and Distant Recurrence-Free Survival With Individual-Level Pathologic Complete Response in Neoadjuvant Treatment of Stages 2 and 3 Breast Cancer: Three-Year Follow-up Analysis for the I-SPY2 Adaptively Randomized Clinical Trial. JAMA Oncol 2020; 6:1355-1362. [PMID: 32701140 PMCID: PMC7378873 DOI: 10.1001/jamaoncol.2020.2535] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/17/2020] [Indexed: 01/04/2023]
Abstract
Importance Pathologic complete response (pCR) is a known prognostic biomarker for long-term outcomes. The I-SPY2 trial evaluated if the strength of this clinical association persists in the context of a phase 2 neoadjuvant platform trial. Objective To evaluate the association of pCR with event-free survival (EFS) and pCR with distant recurrence-free survival (DRFS) in subpopulations of women with high-risk operable breast cancer treated with standard therapy or one of several novel agents. Design, Setting, and Participants Multicenter platform trial of women with operable clinical stage 2 or 3 breast cancer with no prior surgery or systemic therapy for breast cancer; primary tumors were 2.5 cm or larger. Women with tumors that were ERBB2 negative/hormone receptor (HR) positive with low 70-gene assay score were excluded. Participants were adaptively randomized to one of several different investigational regimens or control therapy within molecular subtypes from March 2010 through 2016. The analysis included participants with follow-up data available as of February 26, 2019. Interventions Standard-of-care neoadjuvant therapy consisting of taxane treatment with or without (as control) one of several investigational agents or combinations followed by doxorubicin and cyclophosphamide. Main Outcomes and Measures Pathologic complete response and 3-year EFS and DRFS. Results Of the 950 participants (median [range] age, 49 [23-77] years), 330 (34.7%) achieved pCR. Three-year EFS and DRFS for patients who achieved pCR were both 95%. Hazard ratios for pCR vs non-pCR were 0.19 for EFS (95% CI, 0.12-0.31) and 0.21 for DRFS (95% CI, 0.13-0.34) and were similar across molecular subtypes, varying from 0.14 to 0.18 for EFS and 0.10 to 0.20 for DRFS. Conclusions and Relevance The 3-year outcomes from the I-SPY2 trial show that, regardless of subtype and/or treatment regimen, including 9 novel therapeutic combinations, achieving pCR after neoadjuvant therapy implies approximately an 80% reduction in recurrence rate. The goal of the I-SPY2 trial is to rapidly identify investigational therapies that may improve pCR when validated in a phase 3 confirmatory trial. Whether pCR is a validated surrogate in the sense that a therapy that improves pCR rate can be assumed to also improve long-term outcome requires further study. Trial Registration ClinicalTrials.gov Identifier: NCT01042379.
Collapse
Affiliation(s)
- Douglas Yee
- Masonic Cancer Center, University of Minnesota, Minneapolis
| | | | - Christina Yau
- Department of Surgery, University of California, San Francisco
| | - Claudine Isaacs
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - W Fraser Symmans
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston
| | - Kathy S Albain
- Loyola University Chicago Stritch School of Medicine, Chicago, Illinois
| | - Yunn-Yi Chen
- Department of Pathology, University of California, San Francisco
| | - Gregor Krings
- Department of Pathology, University of California, San Francisco
| | - Shi Wei
- Department of Pathology, University of Alabama Birmingham
| | - Shuko Harada
- Department of Pathology, University of Alabama Birmingham
| | - Brian Datnow
- Department of Pathology, University of California, San Diego
| | - Oluwole Fadare
- Department of Pathology, University of California, San Diego
| | - Molly Klein
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis
| | - Stefan Pambuccian
- Department of Pathology, Loyola University Chicago Stritch School of Medicine, Chicago, Illinois
| | - Beiyun Chen
- Department of Laboratory Medicine and Pathology, Mayo Clinic Rochester, Rochester, Minnesota
| | - Kathi Adamson
- Department of Pathology, Swedish Cancer Institute, Seattle, Washington
| | - Sharon Sams
- Department of Pathology, University of Colorado, Denver
| | | | | | - Mike Feldman
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia
| | - Mara Rendi
- Department of Anatomic Pathology, University of Washington, Seattle
| | - Husain Sattar
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Jay Zeck
- Department of Pathology, Georgetown University, Washington, DC
| | - Idris T Ocal
- Laboratory Medicine and Pathology, Mayo Clinic Scottsdale, Scottsdale, Arizona
| | - Ossama Tawfik
- Department of Pathology and Laboratory Medicine, University of Kansas, Lawrence
| | | | - Sunati Sahoo
- Department of Pathology, University of Texas Southwestern, Dallas
| | - Tuyethoa Vinh
- Inova Pathology Institute, Inova Health System, Falls Church, Virginia
| | - A Jo Chien
- Division of Hematology and Oncology, University of California, San Francisco
| | | | | | - Anne M Wallace
- Department of Surgery, University of California, San Diego
| | - Lajos Pusztai
- Medical Oncology, Yale Cancer Center, New Haven, Connecticut
| | - Judy C Boughey
- Department of Surgery, Mayo Clinic Rochester, Rochester, Minnesota
| | - Erin D Ellis
- Medical Oncology, Swedish Cancer Institute, Seattle, Washington
| | | | - Janice Lu
- Medical Oncology, Keck School of Medicine, University of Southern California, Los Angeles
| | - Julie E Lang
- Surgery, Keck School of Medicine, University of Southern California, Los Angeles
| | - Hyo S Han
- Medical Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Amy S Clark
- Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Rita Nanda
- Hematology and Oncology, University of Chicago Medical Center, Chicago, Illinois
| | | | - Qamar J Khan
- Medical Oncology, University of Kansas Medical Center, Lawrence
| | | | - David M Euhus
- Department of Surgery, Johns Hopkins Medicine, Baltimore, Maryland
| | | | | | - Kathleen Kemmer
- Knight Cancer Institute, Oregon Health & Science University, Portland
| | - John W Park
- Division of Hematology and Oncology, University of California, San Francisco
| | - Minetta C Liu
- Department of Oncology, Mayo Clinic Rochester, Rochester, Minnesota
| | - Olufunmilayo Olopade
- Hematology and Oncology, University of Chicago Medical Center, Chicago, Illinois
| | | | - Debasish Tripathy
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Stacy L Moulder
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Hope S Rugo
- Division of Hematology and Oncology, University of California, San Francisco
| | | | - Shelly Lo
- Medical Oncology, Loyola University Chicago Stritch School of Medicine, Chicago, Illinois
| | | | | | | | - Nola M Hylton
- Department of Radiology and Biomedical Imaging, University of California, San Francisco
| | - Laura J Van't Veer
- Department of Laboratory Medicine, University of California, San Francisco
| | | | - Michelle E Melisko
- Division of Hematology and Oncology, University of California, San Francisco
| | - Amy Wilson
- Quantum Leap Healthcare Collaborative, San Francisco, California
| | - Garry Peterson
- Department of Surgery, University of California, San Francisco
| | - Adam L Asare
- Quantum Leap Healthcare Collaborative, San Francisco, California
| | | | | | | | - Gillian L Hirst
- Department of Surgery, University of California, San Francisco
| | - Ruby Singhrao
- Department of Surgery, University of California, San Francisco
| | - Katherine Steeg
- Department of Surgery, University of California, San Francisco
| | | | - Smita M Asare
- Quantum Leap Healthcare Collaborative, San Francisco, California
| | | | | | | | | |
Collapse
|
24
|
von der Lippe Gythfeldt H, Lien T, Tekpli X, Silwal-Pandit L, Borgen E, Garred Ø, Skjerven H, Schlichting E, Lundgren S, Wist E, Naume B, Kristensen V, Børresen-Dale AL, Lingjaerde OC, Engebraaten O. Immune phenotype of tumor microenvironment predicts response to bevacizumab in neoadjuvant treatment of ER-positive breast cancer. Int J Cancer 2020; 147:2515-2525. [PMID: 32488909 DOI: 10.1002/ijc.33108] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 04/02/2020] [Accepted: 04/30/2020] [Indexed: 12/24/2022]
Abstract
Antiangiogenic drugs are potentially a useful supplement to neoadjuvant chemotherapy for a subgroup of patients with human epidermal growth factor receptor 2 (HER2) negative breast cancer, but reliable biomarkers for improved response are lacking. Here, we report on a randomized phase II clinical trial to study the added effect of bevacizumab in neoadjuvant chemotherapy with FEC100 (5-fluorouracil, epirubicin and cyclophosphamide) and taxanes (n = 132 patients). Gene expression from the tumors was obtained before neoadjuvant treatment, and treatment response was evaluated by residual cancer burden (RCB) at time of surgery. Bevacizumab increased the proportion of complete responders (RCB class 0) from 5% to 20% among patients with estrogen receptor (ER) positive tumors (P = .02). Treatment with bevacizumab was associated with improved 8-year disease-free survival (P = .03) among the good responders (RCB class 0 or I). Patients treated with paclitaxel (n = 45) responded better than those treated with docetaxel (n = 21; P = .03). Improved treatment response was associated with higher proliferation rate and an immune phenotype characterized by high presence of classically activated M1 macrophages, activated NK cells and memory activated CD4 T cells. Treatment with bevacizumab increased the number of adverse events, including hemorrhage, hypertension, infection and febrile neutropenia, but despite this, the ECOG status was not affected.
Collapse
Affiliation(s)
- Hedda von der Lippe Gythfeldt
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Tonje Lien
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Xavier Tekpli
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Laxmi Silwal-Pandit
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Elin Borgen
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Øystein Garred
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Helle Skjerven
- Department of Breast and Endocrine Surgery, Drammen Hospital, Vestre Viken Hospital Trust, Drammen, Norway
| | - Ellen Schlichting
- Department of Breast and Endocrine Surgery, Oslo University Hospital, Oslo, Norway
| | - Steinar Lundgren
- Department of Oncology, St. Olavs University Hospital, Trondheim, Norway
| | - Erik Wist
- Department of Oncology, Oslo University Hospital, Oslo, Norway.,Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Bjørn Naume
- Department of Oncology, Oslo University Hospital, Oslo, Norway.,Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Vessela Kristensen
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Anne-Lise Børresen-Dale
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ole Christian Lingjaerde
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Department of Informatics, University of Oslo, Oslo, Norway.,KG Jebsen Centre for B-Cell Malignancies, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Olav Engebraaten
- Department of Oncology, Oslo University Hospital, Oslo, Norway.,Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
25
|
Diana A, Carlino F, Franzese E, Oikonomidou O, Criscitiello C, De Vita F, Ciardiello F, Orditura M. Early Triple Negative Breast Cancer: Conventional Treatment and Emerging Therapeutic Landscapes. Cancers (Basel) 2020; 12:E819. [PMID: 32235297 PMCID: PMC7225917 DOI: 10.3390/cancers12040819] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 03/19/2020] [Accepted: 03/27/2020] [Indexed: 12/13/2022] Open
Abstract
Triple negative breast cancers (TNBCs) are characterized by worse prognosis, higher propensity to earlier metastases, and shorter survival after recurrence compared with other breast cancer subtypes. Anthracycline- and taxane-based chemotherapy is still the mainstay of treatment in early stages, although several escalation approaches have been evaluated to improve survival outcomes. The addition of platinum salts to standard neoadjuvant chemotherapy (NACT) remains controversial due to the lack of clear survival advantage, and the use of adjuvant capecitabine represents a valid treatment option in TNBC patients with residual disease after NACT. Recently, several clinical trials showed promising results through the use of poly ADP-ribose polymerase (PARP) inhibitors and by incorporating immunotherapy with chemotherapy, enriching treatment options beyond conventional cytotoxic agents. In this review, we provided an overview on the current standard of care and a comprehensive update of the recent advances in the management of early stage TNBC and focused on the latest emerging biomarkers and their clinical application to select the best therapeutic strategy in this hard-to-treat population.
Collapse
Affiliation(s)
- Anna Diana
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy; (F.C.); (E.F.); (F.D.V.); (F.C.); (M.O.)
| | - Francesca Carlino
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy; (F.C.); (E.F.); (F.D.V.); (F.C.); (M.O.)
| | - Elisena Franzese
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy; (F.C.); (E.F.); (F.D.V.); (F.C.); (M.O.)
| | - Olga Oikonomidou
- Cancer Research UK, Edinburgh Centre, MRC Institute Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XR, UK;
| | | | - Ferdinando De Vita
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy; (F.C.); (E.F.); (F.D.V.); (F.C.); (M.O.)
| | - Fortunato Ciardiello
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy; (F.C.); (E.F.); (F.D.V.); (F.C.); (M.O.)
| | - Michele Orditura
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy; (F.C.); (E.F.); (F.D.V.); (F.C.); (M.O.)
| |
Collapse
|
26
|
Vaklavas C, Roberts BS, Varley KE, Lin NU, Liu MC, Rugo HS, Puhalla S, Nanda R, Storniolo AM, Carey LA, Saleh MN, Li Y, Delossantos JF, Grizzle WE, LoBuglio AF, Myers RM, Forero-Torres A. TBCRC 002: a phase II, randomized, open-label trial of preoperative letrozole with or without bevacizumab in postmenopausal women with newly diagnosed stage 2/3 hormone receptor-positive and HER2-negative breast cancer. Breast Cancer Res 2020; 22:22. [PMID: 32070401 PMCID: PMC7027068 DOI: 10.1186/s13058-020-01258-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 02/06/2020] [Indexed: 12/19/2022] Open
Abstract
Background In preclinical studies, the expression of vascular endothelial growth factor (VEGF) in hormone receptor-positive breast cancer is associated with estrogen-independent tumor growth and resistance to endocrine therapies. This study investigated whether the addition of bevacizumab, a monoclonal antibody against VEGF, to letrozole enhanced the antitumor activity of the letrozole in the preoperative setting. Methods Postmenopausal women with newly diagnosed stage 2 or 3 estrogen and/or progesterone receptor-positive, HER2-negative breast cancer were randomly assigned (2:1) between letrozole 2.5 mg PO daily plus bevacizumab 15 mg/kg IV every 3 weeks (Let/Bev) and letrozole 2.5 mg PO daily (Let) for 24 weeks prior to definitive surgery. Primary objective was within-arm pathologic complete remission (pCR) rate. Secondary objectives were safety, objective response, and downstaging rate. Results Seventy-five patients were randomized (Let/Bev n = 50, Let n = 25). Of the 45 patients evaluable for pathological response in the Let/Bev arm, 5 (11%; 95% CI, 3.7–24.1%) achieved pCR and 4 (9%; 95% CI, 2.5–21.2%) had microscopic residual disease; no pCRs or microscopic residual disease was seen in the Let arm (0%; 95% CI, 0–14.2%). The rates of downstaging were 44.4% (95% CI, 29.6–60.0%) and 37.5% (95% CI, 18.8–59.4%) in the Let/Bev and Let arms, respectively. Adverse events typically associated with letrozole (hot flashes, arthralgias, fatigue, myalgias) occurred in similar frequencies in the two arms. Hypertension, headache, and proteinuria were seen exclusively in the Let/Bev arm. The rates of grade 3 and 4 adverse events and discontinuation due to adverse events were 18% vs 8% and 16% vs none in the Let/Bev and Let arms, respectively. A small RNA-based classifier predictive of response to preoperative Let/Bev was developed and confirmed on an independent cohort. Conclusion In the preoperative setting, the addition of bevacizumab to letrozole was associated with a pCR rate of 11%; no pCR was seen with letrozole alone. There was additive toxicity with the incorporation of bevacizumab. Responses to Let/Bev can be predicted from the levels of 5 small RNAs in a pretreatment biopsy. Trial registration This trial is registered with ClinicalTrials.gov (Identifier: NCT00161291), first posted on September 12, 2005, and is completed.
Collapse
Affiliation(s)
- Christos Vaklavas
- University of Alabama at Birmingham, Birmingham, AL, USA.,Present Address: Huntsman Cancer Institute, University of Utah, Salt Lake City, USA
| | - Brian S Roberts
- HudsonAlpha, Institute for Biotechnology, Huntsville, AL, USA
| | | | - Nancy U Lin
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Minetta C Liu
- Lombardi Cancer Center, Georgetown University Hospital, Washington, DC, USA
| | - Hope S Rugo
- University of California, San Francisco, USA.,Hellen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
| | - Shannon Puhalla
- University of Pittsburgh Medical Center, Magee Women's Cancer Program, Pittsburgh, PA, USA
| | | | - Anna Maria Storniolo
- Melvin and Bren Simon Cancer Center at Indiana University, Indianapolis, IN, USA
| | - Lisa A Carey
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
| | | | - Yufeng Li
- University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | - Richard M Myers
- HudsonAlpha, Institute for Biotechnology, Huntsville, AL, USA
| | | | | |
Collapse
|
27
|
Karn T, Meissner T, Weber KE, Solbach C, Denkert C, Engels K, Fasching PA, Sinn BV, Schrader I, Budczies J, Marmé F, Müller V, Holtrich U, Gerber B, Schem C, Young BM, Hanusch C, Stickeler E, Huober J, van Mackelenbergh M, Leyland-Jones B, Fehm T, Nekljudova V, Untch M, Loibl S. A Small Hypoxia Signature Predicted pCR Response to Bevacizumab in the Neoadjuvant GeparQuinto Breast Cancer Trial. Clin Cancer Res 2020; 26:1896-1904. [PMID: 31932495 DOI: 10.1158/1078-0432.ccr-19-1954] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 11/19/2019] [Accepted: 01/08/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE In breast cancer, bevacizumab increased pCR rate but not long-term survival and no predictive markers are available to identify patients with long-term benefit from the drug. EXPERIMENTAL DESIGN We profiled 289 pretherapeutic formalin-fixed, paraffin-embedded (FFPE) biopsies of HER2-negative patients from the GeparQuinto trial of neoadjuvant chemotherapy ± bevacizumab by exome-capture RNA-sequencing (RNA-seq). In a prospectively planned study, we tested molecular signatures for response prediction. IHC validation was performed using tissue microarrays. RESULTS We found strong agreement of molecular and pathologic parameters as hormone receptors, grading, and lymphocyte infiltration in 221 high-quality samples. Response rates (49.3% pCR overall) were higher in basal-like (68.9%) and HER2-enriched (45.5%) than in luminal B (35.7%), luminal A (17.9%), and normal-like (20.0%) subtypes. T-cell (OR = 1.60; 95% confidence interval, 1.21-2.12; P = 0.001), proliferation (OR = 2.88; 95% CI, 2.00-4.15; P < 0.001), and hypoxia signatures (OR = 1.92; 95% CI, 1.41-2.60; P < 0.001) significantly predicted pCR in univariate analysis. In a prespecified multivariate logistic regression, a small hypoxia signature predicted pCR (OR = 2.40; 95% CI, 1.28-4.51; P = 0.006) with a significant interaction with bevacizumab treatment (P = 0.020). IHC validation using NDRG1 as marker revealed highly heterogenous expression within tissue leading to profound loss of sensitivity in TMA analysis, still a significant predictive value for pCR was detected (P = 0.025). CONCLUSIONS Exome-capture RNA-seq characterizes small FFPE core biopsies by reliably detecting factors as for example ER status, grade, and tumor-infiltrating lymphocytes levels. Beside molecular subtypes and immune signatures, a small hypoxia signature predicted pCR to bevacizumab, which could be validated by IHC. The signature can have important applications for bevacizumab treatment in different cancer types and might also have a role for novel combination therapies of bevacizumab with immune checkpoint inhibition.
Collapse
Affiliation(s)
- Thomas Karn
- Goethe University Hospital Frankfurt, Frankfurt, Germany.
| | | | | | | | | | - Knut Engels
- Goethe University Hospital Frankfurt, Frankfurt, Germany
| | - Peter A Fasching
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Germany
| | | | - Iris Schrader
- Gynäkologisch-Onkologische Praxis Hannover, Hannover, Germany
| | | | | | - Volkmar Müller
- University Hospital Hamburg-Eppendorf, Hamburg-Eppendorf, Germany
| | - Uwe Holtrich
- Goethe University Hospital Frankfurt, Frankfurt, Germany
| | | | | | | | | | | | | | | | | | - Tanja Fehm
- University Hospital Tübingen, Tübingen, Germany
| | | | | | | |
Collapse
|
28
|
Banys-Paluchowski M, Reinhardt F, Fehm T. Disseminated Tumor Cells and Dormancy in Breast Cancer Progression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1220:35-43. [PMID: 32304078 DOI: 10.1007/978-3-030-35805-1_3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Hematogenous dissemination of single cancer cells is a common phenomenon in patients with solid tumors. These cells may experience different fates: most will die during the process; some will grow into metastasis and some will persist in secondary homing sites for many years in a state referred to as dormancy. The mechanisms of this state are still not clear; single cancer cells can survive either by completely withdrawing from the cell cycle or by continuing to proliferate at a slow rate that is counterbalanced by cell death. Another hypothesis assumes that at least some of dormant tumor cells feature stem cell-like characteristics that may contribute to their extremely long half-lives and enhance chemotherapy resistance. Breast cancer is particularly known for prolonged periods of clinical freedom of disease (sometimes up to 20-30 years), followed by a distant relapse. In this chapter, we explore the relationship between the clinical phenomenon of tumor dormancy and the disseminated tumor cells and discuss the potential implications for treatment.
Collapse
Affiliation(s)
| | - Florian Reinhardt
- Department of Obstetrics and Gynecology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Tanja Fehm
- Department of Obstetrics and Gynecology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
29
|
Fitzpatrick A, Tutt A. Controversial issues in the neoadjuvant treatment of triple-negative breast cancer. Ther Adv Med Oncol 2019; 11:1758835919882581. [PMID: 31700549 PMCID: PMC6826917 DOI: 10.1177/1758835919882581] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 09/09/2019] [Indexed: 12/12/2022] Open
Abstract
Triple-negative breast cancer (TNBC), as a collective group of heterogenous tumours, displays the highest rate of distant recurrence and lowest survival from metastatic disease across breast cancer subtypes. However, a subset of TNBC display impressive primary tumour response to neoadjuvant chemotherapy, translating to reduction in future relapse and increased overall survival. Maximizing early treatment response is crucial to improving the outlook in this subtype. Numerous systemic therapy strategies are being assessed in the neoadjuvant setting and the current paradigm of generic chemotherapy components in regimens for high-risk breast cancers, regardless of biological subtype, is changing. Therapeutic approaches with evidence of benefit include platinum drugs, polyadenosine diphosphate ribose polymerase (PARP) inhibitors, immunotherapy and second adjuvant therapy for those not achieving pathological complete response. Importantly, molecular testing can identify subgroups within TNBC, such as deoxyribonucleic acid (DNA) homologous recombination repair deficiency, lymphocyte-predominant tumours, and TNBC type 4 molecular subtypes. Clinical trials that address the interaction between these biomarkers and treatment approaches are a priority, to identify subgroups benefiting from additional therapy.
Collapse
Affiliation(s)
- Amanda Fitzpatrick
- Breast Cancer Now Research Centre, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Andrew Tutt
- Breast Cancer Now Research Centre, Institute of Cancer Research, London, UK Faculty of Life Sciences and Medicine, King's College London, London, UK
| |
Collapse
|
30
|
Rounis K, Mavroudis D. A new paradox for pCR in BRCA mutation carriers. Transl Cancer Res 2019; 8:S99-S102. [PMID: 35117073 PMCID: PMC8799287 DOI: 10.21037/tcr.2018.10.25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 10/29/2018] [Indexed: 11/11/2022]
Affiliation(s)
- Konstantinos Rounis
- Department of Medical Oncology, University Hospital of Heraklion, Crete, Greece
| | - Dimitrios Mavroudis
- Department of Medical Oncology, University Hospital of Heraklion, Crete, Greece
| |
Collapse
|
31
|
Wan G, Cao F, Wang X, Sun X. Bevacizumab Added to Neoadjuvant Chemotherapy in HER2-Negative Non-Metastatic Breast Cancer. J Cancer 2019; 10:416-417. [PMID: 30719135 PMCID: PMC6360292 DOI: 10.7150/jca.29461] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Accepted: 11/01/2018] [Indexed: 12/02/2022] Open
Affiliation(s)
- Guoxing Wan
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Fengjun Cao
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Xuanbin Wang
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Xue Sun
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan 442000, China
| |
Collapse
|
32
|
Alnimer Y, Hindi Z, Katato K. The Effect of Perioperative Bevacizumab on Disease-Free and Overall Survival in Locally Advanced HER-2 Negative Breast Cancer: A Meta-Analysis. BREAST CANCER-BASIC AND CLINICAL RESEARCH 2018; 12:1178223418792250. [PMID: 30090017 PMCID: PMC6077892 DOI: 10.1177/1178223418792250] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 07/11/2018] [Indexed: 12/18/2022]
Abstract
Introduction Multiple trials demonstrated that adding Bevacizumab to the standard neoadjuvant chemotherapy in HER-2 negative breast cancer increases pathological complete response. We conducted this meta-analysis to evaluate that effect on survival. Methods We performed a systematic search for randomized trials measuring the effect of adding either neoadjuvant or adjuvant Bevacizumab to the standard chemotherapy on disease-free and overall survival in breast cancer surgical candidates. The Mantel-Haenszel method and random effect model were used to analyze the data. A total of 7 randomized controlled trials were included in the analysis with a mean follow-up of 45 months. Results No statistically significant difference in overall survival was found after adding Bevacizumab to the standard chemotherapy in the overall study population, HR=0.9, 95% CI (90.72-1.13), estrogen/ progesterone positive subgroup, HR=0.99, 95% CI (0.72-1.35), or in triple negative breast cancer, HR=0.88, 95% CI (0.77-1.01). However, there was a small but significant improvement in disease-free survival in triple negative breast cancer with a HR of 0.88, 95% CI (0.78-0.98), but not in estrogen/ progesterone receptor positive tumors, HR=1.01, 95% CI (0.81-1.26). Conclusions The addition of Bevacizumab along with the standard chemotherapy would not improve overall survival in breast cancer surgical candidates, however, due to a small but significant improvement on disease-free survival in triple negative breast cancer, that would not eliminate the possibility of a certain subgroup of the latter who might benefit from adding Bevacizumab.
Collapse
Affiliation(s)
- Yanal Alnimer
- Internal Medicine Department, Hurley Medical Center, Michigan State University, East Lansing, MI, USA
| | - Zakaria Hindi
- Internal Medicine Department, Texas Tech University Health Sciences Center (Permian Basin), Odessa, TX, USA
| | - Khalil Katato
- Hurley Medical Center, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
33
|
Bartsch R, Bergen E, Galid A. Current concepts and future directions in neoadjuvant chemotherapy of breast cancer. MEMO-MAGAZINE OF EUROPEAN MEDICAL ONCOLOGY 2018; 11:199-203. [PMID: 30220926 PMCID: PMC6132793 DOI: 10.1007/s12254-018-0421-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 07/10/2018] [Indexed: 12/21/2022]
Abstract
Preoperative administration of chemotherapy is a widespread treatment approach in early stage breast cancer whenever chemotherapy is indicated in principle. In addition, neoadjuvant treatment is today regarded as the preferred way of delivering systemic therapy in triple-negative and HER2-positive breast cancer. While preoperative chemotherapy allows for disease downstaging and increases breast conservation rates, achieving pathologic complete remission (pCR) is usually regarded as the most pertinent aim as pCR predicts for improved long-term outcome in high-risk breast cancer subtypes. A multitude of clinical trials therefore have focused on strategies to increase pCR rates. This short review summarizes outcomes of selected studies investigating the addition of further chemotherapeutic drugs or biologically targeted agents to standard regimens and provides an overview of novel strategies currently under clinical evaluation.
Collapse
Affiliation(s)
- Rupert Bartsch
- 1Department of Medicine 1, Clinical Division of Oncology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Elisabeth Bergen
- 1Department of Medicine 1, Clinical Division of Oncology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Arik Galid
- 2Department of Gynaecology, Hanusch Hospital, Vienna, Austria
| |
Collapse
|
34
|
McAndrew N, DeMichele A. Neoadjuvant Chemotherapy Considerations in Triple-Negative Breast Cancer. THE JOURNAL OF TARGETED THERAPIES IN CANCER 2018; 7:52-69. [PMID: 29577076 PMCID: PMC5865448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The optimal neoadjuvant chemotherapy (NACT) regimen in triple-negative breast cancer (TNBC) has not been clearly defined. Achieving a pathologic complete response (pCR) provides important prognostic information, and, especially in TNBC, is considered a surrogate endpoint for event-free survival. Thus, many neoadjuvant studies in TNBC focus on this as a primary endpoint, and such information may be used for accelerated US Food and Drug Administration approval. Current controversies in the field include: (1) the role of platinum-based compounds; (2) the optimal chemotherapy backbone; and (3) the benefits of additional therapy after surgery. Conflicting results of 2 major studies adding carboplatin to NACT have highlighted the need to balance potential benefits to disease outcomes against increased toxicity. While the PROGECT study suggests efficacy of a nonanthracycline-containing regimen, this is observational data, and evidence in the form of a clinical trial remains to be seen. Data surrounding optimal taxane use support the use of nab-paclitaxel in place of paclitaxel in limited clinical situations. Although bevacizumab may increase pCR rates, this has not translated into survival benefit. Capecitabine shows promise in patients who have not achieved pCR after NACT. The neoadjuvant setting remains an important model for drug development. This review will focus on the most important and most current neoadjuvant trials in women with TNBC.
Collapse
Affiliation(s)
- Nicholas McAndrew
- Department of Medicine, Division of Hematology/Oncology at the University of Pennsylvania
| | - Angela DeMichele
- Department of Medicine, Division of Hematology/Oncology at the University of Pennsylvania; Also at the Abramson Cancer Center, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania
| |
Collapse
|
35
|
Affiliation(s)
- L Pusztai
- Department of Breast Medical Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, USA
| | - B Szekely
- Department of Breast Medical Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, USA
| | - C Hatzis
- Department of Breast Medical Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, USA
| |
Collapse
|
36
|
Pisarsky L, Ghajar CM. Anti-angiogenic Therapy-Mediated Endothelial Damage: A Driver of Breast Cancer Recurrence? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1100:19-45. [DOI: 10.1007/978-3-319-97746-1_2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
37
|
Gampenrieder SP, Westphal T, Greil R. Antiangiogenic therapy in breast cancer. MEMO-MAGAZINE OF EUROPEAN MEDICAL ONCOLOGY 2017; 10:194-201. [PMID: 29250196 PMCID: PMC5725520 DOI: 10.1007/s12254-017-0362-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 10/23/2017] [Indexed: 12/21/2022]
Abstract
Based on a strong rationale for anti-VEGF (vascular endothelial growth factor) treatment in breast cancer and promising preclinical data, great hopes have been placed on the anti-VEGF antibody bevacizumab. Clinical trials, however, reported conflicting results. In metastatic human epidermal growth factor receptor 2(HER2)-negative breast cancer, the addition of bevacizumab to standard chemotherapy improved consistently progression-free survival (PFS), however, without effect on overall survival (OS). In early breast cancer bevacizumab increased the pathologic complete response rate (pCR) after neoadjuvant therapy, but adjuvant trials did not demonstrate an effect on long-term survival. Unfortunately, despite extensive research, there is still no biomarker for bevacizumab efficacy available, making patient selection difficult. This review summarizes all phase III trials investigating efficacy and toxicity of bevacizumab in early, locally advanced and metastatic breast cancer. It recapitulates the main toxicities, gives an overview on biomarker studies and discusses the role and future aspects of antiangiogenic therapy in breast cancer.
Collapse
Affiliation(s)
- Simon Peter Gampenrieder
- IIIrd Medical Department with Hematology and Medical Oncology, Oncologic Center, Paracelsus Medical University Salzburg, Müllner Hauptstraße 48, 5020 Salzburg, Austria.,Laboratory of Immunological and Molecular Cancer Research and Center for Clinical Cancer and Immunology Trials, Salzburg Cancer Research Institute, Salzburg, Austria.,Cancer Cluster Salzburg, Salzburg, Austria
| | - Theresa Westphal
- IIIrd Medical Department with Hematology and Medical Oncology, Oncologic Center, Paracelsus Medical University Salzburg, Müllner Hauptstraße 48, 5020 Salzburg, Austria.,Laboratory of Immunological and Molecular Cancer Research and Center for Clinical Cancer and Immunology Trials, Salzburg Cancer Research Institute, Salzburg, Austria.,Cancer Cluster Salzburg, Salzburg, Austria
| | - Richard Greil
- IIIrd Medical Department with Hematology and Medical Oncology, Oncologic Center, Paracelsus Medical University Salzburg, Müllner Hauptstraße 48, 5020 Salzburg, Austria.,Laboratory of Immunological and Molecular Cancer Research and Center for Clinical Cancer and Immunology Trials, Salzburg Cancer Research Institute, Salzburg, Austria.,Cancer Cluster Salzburg, Salzburg, Austria
| |
Collapse
|
38
|
Ali HR, Dariush A, Thomas J, Provenzano E, Dunn J, Hiller L, Vallier AL, Abraham J, Piper T, Bartlett JMS, Cameron DA, Hayward L, Brenton JD, Pharoah PDP, Irwin MJ, Walton NA, Earl HM, Caldas C. Lymphocyte density determined by computational pathology validated as a predictor of response to neoadjuvant chemotherapy in breast cancer: secondary analysis of the ARTemis trial. Ann Oncol 2017; 28:1832-1835. [PMID: 28525534 PMCID: PMC5834010 DOI: 10.1093/annonc/mdx266] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND We have previously shown lymphocyte density, measured using computational pathology, is associated with pathological complete response (pCR) in breast cancer. The clinical validity of this finding in independent studies, among patients receiving different chemotherapy, is unknown. PATIENTS AND METHODS The ARTemis trial randomly assigned 800 women with early stage breast cancer between May 2009 and January 2013 to three cycles of docetaxel, followed by three cycles of fluorouracil, epirubicin and cyclophosphamide once every 21 days with or without four cycles of bevacizumab. The primary endpoint was pCR (absence of invasive cancer in the breast and lymph nodes). We quantified lymphocyte density within haematoxylin and eosin (H&E) whole slide images using our previously described computational pathology approach: for every detected lymphocyte the average distance to the nearest 50 lymphocytes was calculated and the density derived from this statistic. We analyzed both pre-treatment biopsies and post-treatment surgical samples of the tumour bed. RESULTS Of the 781 patients originally included in the primary endpoint analysis of the trial, 609 (78%) were included for baseline lymphocyte density analyses and a subset of 383 (49% of 781) for analyses of change in lymphocyte density. The main reason for loss of patients was the availability of digitized whole slide images. Pre-treatment lymphocyte density modelled as a continuous variable was associated with pCR on univariate analysis (odds ratio [OR], 2.92; 95% CI, 1.78-4.85; P < 0.001) and after adjustment for clinical covariates (OR, 2.13; 95% CI, 1.24-3.67; P = 0.006). Increased pre- to post-treatment lymphocyte density showed an independent inverse association with pCR (adjusted OR, 0.1; 95% CI, 0.033-0.31; P < 0.001). CONCLUSIONS Lymphocyte density in pre-treatment biopsies was validated as an independent predictor of pCR in breast cancer. Computational pathology is emerging as a viable and objective means of identifying predictive biomarkers for cancer patients. CLINICALTRIALS.GOV NCT01093235.
Collapse
Affiliation(s)
- H. R. Ali
- Li Ka Shing Centre, Cancer Research UK Cambridge Institute
- Department of Pathology
| | - A. Dariush
- Institute of Astronomy, University of Cambridge, Cambridge
| | - J. Thomas
- Edinburgh Cancer Research Centre, Western General Hospital, Edinburgh
| | - E. Provenzano
- Department of Oncology, University of Cambridge, Addenbrooke’s Hospital, Cambridge
- Department of Histopathology, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge
- Cambridge Experimental Cancer Medicine Centre and NIHR Cambridge Biomedical Research Centre, Cambridge
| | - J. Dunn
- Warwick Clinical Trials Unit, University of Warwick, Coventry, UK
| | - L. Hiller
- Warwick Clinical Trials Unit, University of Warwick, Coventry, UK
| | - A.-L. Vallier
- Department of Oncology, University of Cambridge, Addenbrooke’s Hospital, Cambridge
- Cambridge Experimental Cancer Medicine Centre and NIHR Cambridge Biomedical Research Centre, Cambridge
| | - J. Abraham
- Department of Oncology, University of Cambridge, Addenbrooke’s Hospital, Cambridge
- Cambridge Experimental Cancer Medicine Centre and NIHR Cambridge Biomedical Research Centre, Cambridge
| | - T. Piper
- Edinburgh Cancer Research Centre, Western General Hospital, Edinburgh
| | - J. M. S. Bartlett
- Edinburgh Cancer Research Centre, Western General Hospital, Edinburgh
- Ontario Institute for Cancer Research, Toronto, Canada
| | - D. A. Cameron
- Edinburgh Cancer Research Centre, Western General Hospital, Edinburgh
| | - L. Hayward
- Edinburgh Cancer Research Centre, Western General Hospital, Edinburgh
| | - J. D. Brenton
- Li Ka Shing Centre, Cancer Research UK Cambridge Institute
- Department of Oncology, University of Cambridge, Addenbrooke’s Hospital, Cambridge
- Cambridge Experimental Cancer Medicine Centre and NIHR Cambridge Biomedical Research Centre, Cambridge
| | - P. D. P. Pharoah
- Department of Oncology, University of Cambridge, Addenbrooke’s Hospital, Cambridge
- Cambridge Experimental Cancer Medicine Centre and NIHR Cambridge Biomedical Research Centre, Cambridge
| | - M. J. Irwin
- Institute of Astronomy, University of Cambridge, Cambridge
| | - N. A. Walton
- Institute of Astronomy, University of Cambridge, Cambridge
| | - H. M. Earl
- Department of Oncology, University of Cambridge, Addenbrooke’s Hospital, Cambridge
- Cambridge Experimental Cancer Medicine Centre and NIHR Cambridge Biomedical Research Centre, Cambridge
| | - C. Caldas
- Li Ka Shing Centre, Cancer Research UK Cambridge Institute
- Department of Oncology, University of Cambridge, Addenbrooke’s Hospital, Cambridge
- Cambridge Experimental Cancer Medicine Centre and NIHR Cambridge Biomedical Research Centre, Cambridge
| |
Collapse
|