1
|
Chen X, Huang J, Xie X, Chen L, Lan X, Song L, Bai X, Du C. Apatinib and trastuzumab-based chemotherapy for heavily treated primary trastuzumab-resistant metastatic breast cancer. J Cancer Res Ther 2024; 20:1991-1996. [PMID: 39792408 DOI: 10.4103/jcrt.jcrt_979_24] [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: 05/20/2024] [Accepted: 10/28/2024] [Indexed: 01/12/2025]
Abstract
BACKGROUND The low incidence and poor prognosis primary trastuzumab resistance (PTR) in HER2-positive breast cancer has limited research into possible treatments. Thus, it remains unclear whether this group of patients could benefit from nontargeting HER2 antiangiogenic therapy. PATIENTS AND METHODS We collected the medical data for HER2-positive patients with PTR who received apatinib 250 mg and trastuzumab-based chemotherapy (ATBC) between March 18, 2017, and March 31, 2022. All patients had progressed on ≥2 anti-HER2 treatments, including trastuzumab and small molecular tyrosine kinase inhibitors. We evaluated tumor response and safety profiles to ATBC over a median follow-up time of 34.5 months. RESULTS A total of 198 consecutively HER2-positive metastatic breast cancer patients were reviewed; 20 were PTR and received ATBC. The clinical benefit rate of the total cohort was 55.0%. No patient showed a complete response. The median PFS and overall survival (OS) of the entire cohort was 5.7 months (95% CI 2.9-8.5) and 24.6 months (95% CI 6.9-42.4), respectively. The estimated 2-year survival rate was 46.7% (95% CI 38.4-81.6%). The most common nonhematologic adverse events were hypertension (70.0%), hand-foot skin reaction (55.0%), proteinuria (40.0%), and cardiovascular decrease of LVEF (20.0%). No new toxicities were observed. CONCLUSION ATBC had favorable effects for PTR breast cancer patients in later line treatment. The toxicity of the triple-combination regimen was tolerable; thus, further research should focus on identifying PTR patients who could benefit from ATBC.
Collapse
Affiliation(s)
- Xuelian Chen
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
2
|
Nandi D, Sharma D. Integrating immunotherapy with conventional treatment regime for breast cancer patients- an amalgamation of armamentarium. Front Immunol 2024; 15:1477980. [PMID: 39555066 PMCID: PMC11563812 DOI: 10.3389/fimmu.2024.1477980] [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: 08/08/2024] [Accepted: 10/08/2024] [Indexed: 11/19/2024] Open
Abstract
Immunotherapy stands as the frontrunner in treatment strategies imparting efficient remission in various types of cancer. In fact, emerging breakthroughs with immune checkpoint inhibitors (ICI) in a spectrum of cancers have evoked interest in research related to the potential effects of immunotherapy in breast cancer patients. A major challenge with breast cancer is the molecular heterogeneity that limits the efficacy of many therapeutic regimes. Clinical trials have shown favorable clinical outcomes with immunotherapeutic options in some subtypes of breast cancer. However, ICI monotherapy may not be sufficient for all breast cancer patients, emphasizing the need for combinatorial approaches. Ongoing research is focused on untangling the interplay of ICI with established as well as novel anticancer therapeutic regimens in preclinical models of breast cancer. Our review will analyze the existing research regarding the mechanisms and clinical impact of immunotherapy for the treatment of breast cancer. We shall evaluate the role of immune cell modulation for improved therapeutic response in breast cancer patients. This review will provide collated evidences about the current clinical trials that are testing out the implications of immunotherapy in conjunction with traditional treatment modalities in breast cancer and summarize the potential future research directions in the field. In addition, we shall underline the recent findings related to microbiota modulation as a key regulator of immune therapy response in cancer patients and its plausible applications in breast cancer.
Collapse
Affiliation(s)
- Deeptashree Nandi
- Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, United States
| | - Dipali Sharma
- Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, United States
| |
Collapse
|
3
|
Lagarde CB, Thapa K, Cullen NM, Hawes ML, Salim K, Benz MC, Dietrich SR, Burow BE, Bunnell BA, Martin EC, Collins-Burow BM, Lynch RM, Hoang VT, Burow ME, Fang JS. Obesity and leptin in breast cancer angiogenesis. Front Endocrinol (Lausanne) 2024; 15:1465727. [PMID: 39439572 PMCID: PMC11493622 DOI: 10.3389/fendo.2024.1465727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 09/04/2024] [Indexed: 10/25/2024] Open
Abstract
At the time of breast cancer diagnosis, most patients meet the diagnostic criteria to be classified as obese or overweight. This can significantly impact patient outcome: breast cancer patients with obesity (body mass index > 30) have a poorer prognosis compared to patients with a lean BMI. Obesity is associated with hyperleptinemia, and leptin is a well-established driver of metastasis in breast cancer. However, the effect of hyperleptinemia on angiogenesis in breast cancer is less well-known. Angiogenesis is an important process in breast cancer because it is essential for tumor growth beyond 1mm3 in size as well as cancer cell circulation and metastasis. This review investigates the role of leptin in regulating angiogenesis, specifically within the context of breast cancer and the associated tumor microenvironment in obese patients.
Collapse
Affiliation(s)
- Courtney B. Lagarde
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane University Cancer Center, New Orleans, LA, United States
| | - Kapil Thapa
- Department of Cell and Molecular Biology, Tulane University School of Science and Engineering, New Orleans, LA, United States
| | - Nicole M. Cullen
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane University Cancer Center, New Orleans, LA, United States
| | - Mackenzie L. Hawes
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane University Cancer Center, New Orleans, LA, United States
| | - Khudeja Salim
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane University Cancer Center, New Orleans, LA, United States
| | - Megan C. Benz
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane University Cancer Center, New Orleans, LA, United States
| | - Sophie R. Dietrich
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane University Cancer Center, New Orleans, LA, United States
- United States Department of Agriculture Southern Regional Research Center, New Orleans, LA, United States
| | - Brandon E. Burow
- Department of Cell and Molecular Biology, Tulane University School of Science and Engineering, New Orleans, LA, United States
| | - Bruce A. Bunnell
- Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Elizabeth C. Martin
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane University Cancer Center, New Orleans, LA, United States
| | - Bridgette M. Collins-Burow
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane University Cancer Center, New Orleans, LA, United States
| | - Ronald M. Lynch
- Department of Physiology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Van T. Hoang
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane University Cancer Center, New Orleans, LA, United States
| | - Matthew E. Burow
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane University Cancer Center, New Orleans, LA, United States
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, United States
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, United States
| | - Jennifer S. Fang
- Department of Cell and Molecular Biology, Tulane University School of Science and Engineering, New Orleans, LA, United States
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA, United States
| |
Collapse
|
4
|
Guo Z, Jing X, Sun X, Sun S, Yang Y, Cao Y. Tumor angiogenesis and anti-angiogenic therapy. Chin Med J (Engl) 2024; 137:2043-2051. [PMID: 39051171 PMCID: PMC11374217 DOI: 10.1097/cm9.0000000000003231] [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: 12/24/2023] [Indexed: 07/27/2024] Open
Abstract
ABSTRACT Anti-angiogenic drugs (AADs), which mainly target the vascular endothelial growth factor-A signaling pathway, have become a therapeutic option for cancer patients for two decades. During this period, tremendous clinical experience of anti-angiogenic therapy has been acquired, new AADs have been developed, and the clinical indications for AAD treatment of various cancers have been expanded using monotherapy and combination therapy. However, improvements in the therapeutic outcomes of clinically available AADs and the development of more effective next-generation AADs are still urgently required. This review aims to provide historical and perspective views on tumor angiogenesis to allow readers to gain mechanistic insights and learn new therapeutic development. We revisit the history of concept initiation and AAD discovery, and summarize the up-to-date clinical translation of anti-angiogenic cancer therapy in this field.
Collapse
Affiliation(s)
- Ziheng Guo
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xu Jing
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm 17177, Sweden
| | - Xiaoting Sun
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm 17177, Sweden
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vison and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Shishuo Sun
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm 17177, Sweden
- Cancer Institute, the First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Yunlong Yang
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm 17177, Sweden
| |
Collapse
|
5
|
Mezzanotte-Sharpe J, ONeill A, Mayer IA, Arteaga CL, Yang XJ, Wagner LI, Cella D, Meropol NJ, Alpaugh RK, Saphner TJ, Swaney RE, Hoelzer KL, Gradishar WJ, Abramson VG, Sundaram PK, Jilani SZ, Perez EA, Lin NU, Jahanzeb M, Wolff AC, Sledge GW, Reid SA. A randomized phase III double-blind placebo-controlled trial of first-line chemotherapy and trastuzumab with or without bevacizumab for patients with HER2/neu-positive metastatic breast cancer: a trial of the ECOG-ACRIN Cancer Research Group (E1105). Breast Cancer Res Treat 2024; 207:275-282. [PMID: 38967884 PMCID: PMC11297090 DOI: 10.1007/s10549-024-07417-4] [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: 04/19/2024] [Accepted: 06/23/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND In 2008, bevacizumab received accelerated Food and Drug Administration (FDA) approval for use in human epidermal growth factor receptor 2 (HER2)-negative metastatic breast cancer (MBC). Based on the pre-clinical and preliminary clinical activity of the trastuzumab and bevacizumab combination, ECOG-ACRIN E1105 trial was developed to determine if the addition of bevacizumab to a chemotherapy and trastuzumab combination for first-line therapy would improve progression-free survival (PFS) in patients with HER2-positive MBC. FINDINGS 96 patients were randomized to receive standard first-line chemotherapy and trastuzumab with or without bevacizumab between November 2007 and October 2009, and 93 began protocol therapy. Induction therapy was given for 24 weeks, followed by maintenance trastuzumab with or without bevacizumab. 60% (56/93) began carboplatin and 74% (69/93) completed 6 cycles of induction therapy. Primary endpoint was PFS. Median PFS was 11.1 and 13.8 months for placebo and bevacizumab arms, respectively (hazard ratio [HR] 95%, Confidence Interval [Cl] for bevacizumab vs. placebo: 0.73 [0.43-1.23], p = 0.24), and at a median follow-up of 70.7 months, median survival was 49.1 and 63 months (HR [95% Cl] for OS: 1.09 [0.61-1.97], p = 0.75). The most common toxicities across both arms were neutropenia and hypertension, with left ventricular systolic dysfunction, fatigue, and sensory neuropathy reported more frequently with bevacizumab. CONCLUSIONS In this trial, the addition of bevacizumab did not improve outcomes in patients with metastatic HER2-positive breast cancer. Although the trial was underpowered due to smaller than anticipated sample size, these findings corroborated other clinical trials during this time. CLINICAL TRIAL INFORMATION NCT00520975.
Collapse
Affiliation(s)
| | - Anne ONeill
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ingrid A Mayer
- Vanderbilt University Medical Center, 2220 Pierce Avenue, 777 PRB, Nashville, TN, 37232-6307, USA
| | | | | | - Lynne I Wagner
- Wake Forest University Health Sciences, Winston Salem, NC, USA
| | | | | | | | | | | | | | | | - Vandana G Abramson
- Vanderbilt University Medical Center, 2220 Pierce Avenue, 777 PRB, Nashville, TN, 37232-6307, USA
| | | | | | | | - Nancy U Lin
- Dana-Farber Cancer Institute, Boston, MA, USA
| | | | | | | | - Sonya A Reid
- Vanderbilt University Medical Center, 2220 Pierce Avenue, 777 PRB, Nashville, TN, 37232-6307, USA.
| |
Collapse
|
6
|
Li L, Wu Y, Lan B, Ma F. Efficacy and safety of first-line regimens for advanced HER2-positive breast cancer: A Bayesian network meta-analysis. CANCER INNOVATION 2024; 3:e126. [PMID: 38948247 PMCID: PMC11212280 DOI: 10.1002/cai2.126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/23/2024] [Accepted: 03/02/2024] [Indexed: 07/02/2024]
Abstract
Background The current standard of care for advanced human epidermal growth factor receptor 2 (HER2)-positive breast cancer is pertuzumab plus trastuzumab and docetaxel as first-line therapy. However, with the development of newer treatment regimens, there is a lack of evidence regarding which is the optimal treatment strategy. The aim of this network meta-analysis was to evaluate the efficacy and safety of first-line regimens for advanced HER2-positive breast cancer by indirect comparisons. Methods A systematic review and Bayesian network meta-analysis were conducted. The PubMed, EMBASE, and Cochrane Library databases were searched for relevant articles published through to December 2023. The hazard ratio (HR) and 95% credible interval (CrI) were used to compare progression-free survival (PFS) between treatments, and the odds ratio and 95% CrI were used to compare the objective response rate (ORR) and safety. Results Twenty randomized clinical trials that included 15 regimens and 7094 patients were analyzed. Compared with the traditional trastuzumab and docetaxel regimen, PFS was longer on the pyrotinib and trastuzumab plus docetaxel regimen (HR: 0.41, 95% CrI: 0.22-0.75) and the pertuzumab and trastuzumab plus docetaxel regimen (HR: 0.65, 95% CrI: 0.43-0.98). Consistent with the results for PFS, the ORR was better on the pyrotinib and trastuzumab plus docetaxel regimen and the pertuzumab and trastuzumab plus docetaxel regimen than on the traditional trastuzumab and docetaxel regimen. The surface under the cumulative ranking curve indicated that the pyrotinib and trastuzumab plus docetaxel regimen was most likely to rank first in achieving the best PFS and ORR. Comparable results were found for grade ≥3 AE rates of ≥10%. Conclusions Our results suggest that the pyrotinib and trastuzumab plus docetaxel regimen is most likely to be the optimal first-line therapy for patients with HER2-positive breast cancer.
Collapse
Affiliation(s)
- Lixi Li
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yun Wu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Bo Lan
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Fei Ma
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
7
|
Akinpelu A, Akinsipe T, Avila LA, Arnold RD, Mistriotis P. The impact of tumor microenvironment: unraveling the role of physical cues in breast cancer progression. Cancer Metastasis Rev 2024; 43:823-844. [PMID: 38238542 PMCID: PMC11156564 DOI: 10.1007/s10555-024-10166-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 01/02/2024] [Indexed: 01/30/2024]
Abstract
Metastasis accounts for the vast majority of breast cancer-related fatalities. Although the contribution of genetic and epigenetic modifications to breast cancer progression has been widely acknowledged, emerging evidence underscores the pivotal role of physical stimuli in driving breast cancer metastasis. In this review, we summarize the changes in the mechanics of the breast cancer microenvironment and describe the various forces that impact migrating and circulating tumor cells throughout the metastatic process. We also discuss the mechanosensing and mechanotransducing molecules responsible for promoting the malignant phenotype in breast cancer cells. Gaining a comprehensive understanding of the mechanobiology of breast cancer carries substantial potential to propel progress in prognosis, diagnosis, and patient treatment.
Collapse
Affiliation(s)
- Ayuba Akinpelu
- Department of Chemical Engineering, Samuel Ginn College of Engineering, Auburn University, Auburn, AL, 36849, USA
| | - Tosin Akinsipe
- Department of Biological Sciences, College of Science and Mathematics, Auburn University, Auburn, AL, 36849, USA
| | - L Adriana Avila
- Department of Biological Sciences, College of Science and Mathematics, Auburn University, Auburn, AL, 36849, USA
| | - Robert D Arnold
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL, 36849, USA
| | - Panagiotis Mistriotis
- Department of Chemical Engineering, Samuel Ginn College of Engineering, Auburn University, Auburn, AL, 36849, USA.
| |
Collapse
|
8
|
Yoon SM, Bazan JG. Navigating Breast Cancer Oligometastasis and Oligoprogression: Current Landscape and Future Directions. Curr Oncol Rep 2024; 26:647-664. [PMID: 38652425 PMCID: PMC11168988 DOI: 10.1007/s11912-024-01529-2] [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] [Accepted: 03/25/2024] [Indexed: 04/25/2024]
Abstract
PURPOSE We examine the potential for curative approaches among metastatic breast cancer (MBC) patients by exploring the recent literature on local ablative therapies like surgery and stereotactic body radiation therapy (SBRT) in patients with oligometastatic (OM) breast cancer. We also cover therapies for MBC patients with oligoprogressive (OP) disease. KEY FINDINGS Surgery and SBRT have been studied for OM and OP breast cancer, mainly in retrospective or non-randomized trials. While many studies demonstrated favorable results, a cooperative study and single-institution trial found no support for surgery/SBRT in OM and OP cases, respectively. CONCLUSION While there is interest in applying local therapies to OM and OP breast cancer, the current randomized data does not back the routine use of surgery or SBRT, particularly when considering the potential for treatment-related toxicities. Future research should refine patient selection through advanced imaging and possibly explore these therapies specifically in patients with hormone receptor-positive or HER2-positive disease.
Collapse
Affiliation(s)
- Stephanie M Yoon
- Department of Radiation Oncology, City of Hope Comprehensive Cancer Center, 1500 E. Duarte Road, Duarte, CA, 91010, USA
| | - Jose G Bazan
- Department of Radiation Oncology, City of Hope Comprehensive Cancer Center, 1500 E. Duarte Road, Duarte, CA, 91010, USA.
| |
Collapse
|
9
|
Mezzanotte-Sharpe J, ONeill A, Mayer IA, Arteaga CL, Yang XJ, Wagner LI, Cella D, Meropol NJ, Alpaugh RK, Saphner TJ, Swaney RE, Hoelzer KL, Gradishar WJ, Abramson VG, Sundaram PK, Jilani SZ, Perez EA, Lin NU, Jahanzeb M, Wolff AC, Sledge GW, Reid SA. A randomized phase III double-blind placebo-controlled trial of first line chemotherapy and trastuzumab with or without bevacizumab for patients with HER2/neu-positive metastatic breast cancer: a trial of the ECOG-ACRIN Cancer Research Group (E1105). RESEARCH SQUARE 2024:rs.3.rs-4295044. [PMID: 38746356 PMCID: PMC11092826 DOI: 10.21203/rs.3.rs-4295044/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Background In 2008, bevacizumab received accelerated Food and Drug Administration (FDA) approval for use in human epidermal growth factor receptor 2 (HER2)-negative metastatic breast cancer (MBC). Based on the preclinical and preliminary clinical activity of the trastuzumab and bevacizumab combination, ECOG-ACRIN E1105 trial was developed to determine if the addition of bevacizumab to a chemotherapy and trastuzumab combination for first-line therapy would improve progression-free survival (PFS) in patients with HER2-positive MBC. Findings 96 patients were randomized to receive standard first-line chemotherapy and trastuzumab with or without bevacizumab between November 2007 and October 2009, and 93 began protocol therapy. Induction therapy was given for 24 weeks, followed by maintenance trastuzumab with or without bevacizumab. 60% (56/93) began carboplatin and 74% (69/93) completed 6 cycles of induction therapy. Primary endpoint was PFS. Median PFS was 11.1 and 13.8 months for placebo and bevacizumab arms, respectively (hazard ratio [HR] 95%, Confidence Interval [Cl] for bevacizumab vs. placebo: 0.73 [0.43-1.23], p = 0.24), and at a median follow-up of 70.7 months, median survival was 49.1 and 63 months (HR [95% Cl] for OS: 1.09 [0.61-1.97], p = 0.75). The most common toxicities across both arms were neutropenia and hypertension, with left ventricular systolic dysfunction, fatigue, and sensory neuropathy reported more frequently with bevacizumab. Conclusions In this trial, the addition of bevacizumab did not improve outcomes in patients with metastatic HER2-positive breast cancer. Although the trial was underpowered due to smaller than anticipated sample size, these findings corroborated other clinical trials during this time.
Collapse
|
10
|
Wang J, Yu Y, Lin Q, Zhang J, Song C. Efficacy and safety of first-line therapy in patients with HER2-positive advanced breast cancer: a network meta-analysis of randomized controlled trials. J Cancer Res Clin Oncol 2024; 150:21. [PMID: 38244085 PMCID: PMC10799814 DOI: 10.1007/s00432-023-05530-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 11/03/2023] [Indexed: 01/22/2024]
Abstract
PURPOSE The numerous first-line treatment regimens for human epidermal growth factor receptor 2 (HER2)-positive advanced breast cancer (ABC) necessitate a comprehensive evaluation to inform clinical decision-making. We conducted a Bayesian network meta-analysis (NMA) to compare the efficacy and safety of different interventions. METHODS We systematically searched for relevant randomized controlled trials (RCTs) in Pubmed, Embase, Cochrane Library and online abstracts from inception to June 1, 2023. NMA was performed to calculate and analyze progression-free survival (PFS), overall survival (OS), objective response rate (ORR), and adverse events of grade 3 or higher (≥ 3 AEs). RESULTS Out of the 10,313 manuscripts retrieved, we included 28 RCTs involving 11,680 patients. Regarding PFS and ORR, the combination of trastuzumab with tyrosine kinase inhibitors (TKIs) was more favorable than dual-targeted therapy. If only using trastuzumab, combination chemotherapy is superior to monochemotherapy in terms of PFS. It is important to note that the addition of anthracycline did not result in improved PFS. For patients with hormone receptor-positive HER2-positive diseases, dual-targeted combined with endocrine therapy showed better benefit in terms of PFS compared to dual-targeted alone, but it did not reach statistical significance. The comprehensive analysis of PFS and ≥ 3 AEs indicates that monochemotherapy combined with dual-targeted therapy still has the optimal balance between efficacy and safety. CONCLUSION Monochemotherapy (Docetaxel) plus dual-target (Trastuzumab and Pertuzumab) therapy remains the optimal choice among all first-line treatment options for ABC. The combination of trastuzumab with TKIs (Pyrotinib) demonstrated a significant improvement in PFS and ORR, but further data are warranted to confirm the survival benefit.
Collapse
Affiliation(s)
- Junxiao Wang
- Department of Breast Surgery, College of Clinical Medicine for Oncology, Fujian Medical University, No.91, Fuma Road, Jin'an District, Fuzhou, Fujian Province, China
- Department of Thyroid and Breast Surgery, The Second Hospital of Sanming, Sanming City, Fujian Province, China
| | - Yushuai Yu
- Department of Breast Surgery, College of Clinical Medicine for Oncology, Fujian Medical University, No.91, Fuma Road, Jin'an District, Fuzhou, Fujian Province, China
- Breast Surgery Institute, College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Qisheng Lin
- Department of Thyroid and Breast Surgery, The Second Hospital of Sanming, Sanming City, Fujian Province, China
| | - Jie Zhang
- Department of Breast Surgery, Fujian Medical University Union Hospital, No.29, Xin Quan Road, Gulou District, Fuzhou, 350001, Fujian Province, China.
- Breast Surgery Institute, Fujian Medical University, Fuzhou, Fujian Province, China.
| | - Chuangui Song
- Department of Breast Surgery, College of Clinical Medicine for Oncology, Fujian Medical University, No.91, Fuma Road, Jin'an District, Fuzhou, Fujian Province, China.
- Breast Surgery Institute, College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou, Fujian Province, China.
| |
Collapse
|
11
|
Jacobs F, Molinelli C, Martins-Branco D, Marta GN, Salmon M, Ameye L, Piccart M, Lambertini M, Agostinetto E, de Azambuja E. Progression-free survival assessment by local investigators versus blinded independent central review in randomized clinical trials in metastatic breast cancer: A systematic review and meta-analysis. Eur J Cancer 2024; 197:113478. [PMID: 38103328 DOI: 10.1016/j.ejca.2023.113478] [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: 09/20/2023] [Revised: 12/02/2023] [Accepted: 12/04/2023] [Indexed: 12/19/2023]
Abstract
INTRODUCTION In randomized clinical trials (RCTs), blinded independent central review (BICR) is used to minimize heterogeneity and bias associated with radiological response evaluation by local investigators. However, BICR adds costs and complexity to the trial management. We assessed the discrepancy index between progression-free survival (PFS) assessment by local investigators and by BICR in RCTs conducted in patients with metastatic breast cancer (MBC). METHODS A systematic search of PubMed, Embase, Cochrane databases and conference proceedings (ASCO, SABCS, ESMO) was performed up to January 4, 2023 (PROSPERO: CRD42021229865). All RCTs published from 2000 to 2022, including MBC patients treated in first- or second-line, and reporting PFS assessed by local investigators and BICR were included. A discrepancy index between BICR-assessed and investigator-assessed HR was calculated for each trial and an overall combined DI was obtained using a fixed-effects model. The agreement between hazard ratios (HR) of PFS assessed by local investigators and BICR was measured using intraclass correlation coefficient (ICC). RESULTS We analyzed 24 studies including 13,168 patients. Among them, 19 (79%) were in first-line, 18 (75%) were phase III trials and 23 (96%) had PFS as primary endpoint. The overall combined discrepancy index was 0.97 (95%CI 0.85-1.10; ICC 0.831, p < 0.001) suggesting no statistically significant difference in PFS assessment between local investigators and BICR. This result was consistent across all analyzed subgroups. CONCLUSIONS The good concordance between local investigator and BICR assessments supports the reliability of local investigator-assessed PFS as primary endpoint for RCTs in MBC and questions the practical utility of implementing BICR in all RCTs.
Collapse
Affiliation(s)
- Flavia Jacobs
- Université Libre de Bruxelles (U.L.B), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Academic Trials Promoting Team (ATPT), Brussels, Belgium; Humanitas Clinical and Research Center - IRCCS, Humanitas Cancer Center, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Chiara Molinelli
- Université Libre de Bruxelles (U.L.B), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Academic Trials Promoting Team (ATPT), Brussels, Belgium; Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genoa, Italy
| | - Diogo Martins-Branco
- Université Libre de Bruxelles (U.L.B), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Academic Trials Promoting Team (ATPT), Brussels, Belgium
| | - Guilherme Nader Marta
- Université Libre de Bruxelles (U.L.B), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Academic Trials Promoting Team (ATPT), Brussels, Belgium
| | - Maurine Salmon
- Université Libre de Bruxelles (U.L.B), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Data Center, Brussels, Belgium
| | - Lieveke Ameye
- Université Libre de Bruxelles (U.L.B), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Data Center, Brussels, Belgium
| | - Martine Piccart
- Université Libre de Bruxelles (U.L.B), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Medical Oncology Department, Brussels, Belgium
| | - Matteo Lambertini
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genoa, Italy; Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Elisa Agostinetto
- Université Libre de Bruxelles (U.L.B), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Academic Trials Promoting Team (ATPT), Brussels, Belgium.
| | - Evandro de Azambuja
- Université Libre de Bruxelles (U.L.B), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Academic Trials Promoting Team (ATPT), Brussels, Belgium.
| |
Collapse
|
12
|
Ye F, Dewanjee S, Li Y, Jha NK, Chen ZS, Kumar A, Vishakha, Behl T, Jha SK, Tang H. Advancements in clinical aspects of targeted therapy and immunotherapy in breast cancer. Mol Cancer 2023; 22:105. [PMID: 37415164 PMCID: PMC10324146 DOI: 10.1186/s12943-023-01805-y] [Citation(s) in RCA: 138] [Impact Index Per Article: 69.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/08/2023] [Indexed: 07/08/2023] Open
Abstract
Breast cancer is the second leading cause of death for women worldwide. The heterogeneity of this disease presents a big challenge in its therapeutic management. However, recent advances in molecular biology and immunology enable to develop highly targeted therapies for many forms of breast cancer. The primary objective of targeted therapy is to inhibit a specific target/molecule that supports tumor progression. Ak strain transforming, cyclin-dependent kinases, poly (ADP-ribose) polymerase, and different growth factors have emerged as potential therapeutic targets for specific breast cancer subtypes. Many targeted drugs are currently undergoing clinical trials, and some have already received the FDA approval as monotherapy or in combination with other drugs for the treatment of different forms of breast cancer. However, the targeted drugs have yet to achieve therapeutic promise against triple-negative breast cancer (TNBC). In this aspect, immune therapy has come up as a promising therapeutic approach specifically for TNBC patients. Different immunotherapeutic modalities including immune-checkpoint blockade, vaccination, and adoptive cell transfer have been extensively studied in the clinical setting of breast cancer, especially in TNBC patients. The FDA has already approved some immune-checkpoint blockers in combination with chemotherapeutic drugs to treat TNBC and several trials are ongoing. This review provides an overview of clinical developments and recent advancements in targeted therapies and immunotherapies for breast cancer treatment. The successes, challenges, and prospects were critically discussed to portray their profound prospects.
Collapse
Affiliation(s)
- Feng Ye
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India
| | - Yuehua Li
- Department of Medical Oncology, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
- School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, 144411, India
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, 11439, USA
| | - Ankush Kumar
- Pharmaceutical and Health Sciences, Career Point University, Hamirpur, Himachal Pradesh, India
| | - Vishakha
- Pharmaceutical and Health Sciences, Career Point University, Hamirpur, Himachal Pradesh, India
| | - Tapan Behl
- School of Health Sciences and Technology, University of Petroleum and Energy Studies, Bidholi, Dehradun, Uttarakhand, India.
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India.
- Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, 140413, India.
- Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun, 248007, India.
| | - Hailin Tang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
13
|
Wang Y, Xu H, Han Y, Wu Y, Sa Q, Wang J. Identifying the optimal therapeutics for patients with hormone receptor-positive, HER2-positive advanced breast cancer: a systematic review and network meta-analysis. ESMO Open 2023; 8:101216. [PMID: 37084609 PMCID: PMC10172889 DOI: 10.1016/j.esmoop.2023.101216] [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: 02/09/2023] [Revised: 03/12/2023] [Accepted: 03/18/2023] [Indexed: 04/23/2023] Open
Abstract
INTRODUCTION Hormone receptor-positive (HR+) and human epidermal growth factor receptor 2-positive (HER2+) breast cancer is a distinct subtype with different prognosis and response to treatment. HER2-targeted therapy is currently recommended for patients with HR+/HER2+ advanced breast cancer. However, there is debate over which drugs to add on the basis of HER2 blockade yield the optimal efficacy. This systematic review and network meta-analysis was conducted to solve the problem. METHODS Eligible randomized controlled trials (RCTs) comparing different interventions in HR+/HER2+ metastatic breast cancer were included. The outcomes of interest included progression-free survival (PFS), overall survival (OS) and treatment-related adverse events (TRAEs). Pooled hazard ratios or odds ratios with credible intervals (CrIs) were calculated to estimate the predefined outcomes. The optimal therapeutics were identified by comparing the surface under the cumulative ranking curves (SUCRA). RESULTS Totally, 23 literatures of 20 RCTs were included. Regarding PFS, significant differences were detected between single or dual HER2 blockade plus endocrine therapy (ET) versus ET alone and dual HER2 blockade plus ET versus physician's choice. Trastuzumab, pertuzumab plus chemotherapy significantly improved PFS than trastuzumab plus chemotherapy (hazard ratio 0.69, 95% CrI 0.50-0.92). The SUCRA values suggested the relatively better efficacy of dual HER2-targeted therapy plus ET (86%-91%) than chemotherapy (62%-81%) in prolonging PFS and OS. The HER2 blockade-containing regimens showed similar safety profiles in eight documented TRAEs. CONCLUSIONS Prominent status of dual-targeted therapy for patients with HR+/HER2+ metastatic breast cancer was revealed. Compared with chemotherapy-containing regimens, the ET-containing ones showed better efficacy and similar safety profiles, which could be recommended in clinical practice.
Collapse
Affiliation(s)
- Y Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - H Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Y Han
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Y Wu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Q Sa
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - J Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
14
|
Leblanc D, Cantin G, Desnoyers A, Dufresne J, Masucci GL, Panet-Raymond V, Poirier É, Soldera S, Gingras I. Management of Oligometastatic Breast Cancer: An Expert Committee's Opinion. Curr Oncol 2023; 30:1416-1425. [PMID: 36826069 PMCID: PMC9954938 DOI: 10.3390/curroncol30020108] [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: 12/06/2022] [Revised: 01/09/2023] [Accepted: 01/11/2023] [Indexed: 01/20/2023] Open
Abstract
Patients with oligometastatic breast cancer (BC) are candidates of choice for metastasis-directed therapy (MDT). This paper summarizes the opinions of an expert committee about the management of oligometastatic BC. The experts could complete the questionnaire from 13 September 2021, to 10 October 2021, followed by a discussion. The experts were physicians working in the Province of Quebec (Canada) and specialized in BC care, including surgical oncologists, medical oncologists, and radiation oncologists. The experts provided their opinions about the context of the disease and therapeutic approach, local and systemic therapies, and the prognosis of oligometastatic BC. In addition to the expert panel's opinions about the management of oligometastatic disease per se, the experts stated that a prospective data registry should be implemented to collect data about oligometastatic BC to improve knowledge about oligometastatic BC and implement data-driven MDT. These data could also allow for the design of treatment algorithms. In conclusion, this paper presents the expert panel's opinions about the management of oligometastatic BC and highlights the needs to be met to improve the care of this condition.
Collapse
Affiliation(s)
- Dominique Leblanc
- Centre Hospitalier Universitaire de Québec—Université Laval, Québec, QC G1V 0A6, Canada
- Correspondence:
| | - Guy Cantin
- Centre Hospitalier Universitaire de Québec—Université Laval, Québec, QC G1V 0A6, Canada
| | - Alexandra Desnoyers
- Centre Hospitalier Universitaire de Québec—Université Laval, Québec, QC G1V 0A6, Canada
| | - Jean Dufresne
- Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | | | | | - Éric Poirier
- Centre Hospitalier Universitaire de Québec—Université Laval, Québec, QC G1V 0A6, Canada
| | - Sara Soldera
- Hôpital Charles-Le Moyne, Greenfield Park, QC J4V 2H1, Canada
| | | |
Collapse
|
15
|
Katipally RR, Olson AC, Chmura SJ. Flipping the Null: Lessons From Breast Cancer and Recalibrating Expectations in Oligometastases. Int J Radiat Oncol Biol Phys 2022; 114:840-842. [DOI: 10.1016/j.ijrobp.2022.09.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022]
|
16
|
Chow CY, Lie EF, Wu CH, Chow LW. Clinical implication of genetic composition and molecular mechanism on treatment strategies of HER2-positive breast cancers. Front Oncol 2022; 12:964824. [PMID: 36387174 PMCID: PMC9659858 DOI: 10.3389/fonc.2022.964824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 10/14/2022] [Indexed: 12/01/2022] Open
Abstract
The current clinical management model of HER2-positive breast cancers is commonly based on guidelines, which in turn are based on the design and outcome of clinical trials. While this model is useful to most practicing clinicians, the treatment outcome of individual patient is not certain at the start of treatment. As the understanding of the translational research of carcinogenesis and the related changes in cancer genetics and tumor microenvironment during treatment is critical in the selection of right choice of treatment to maximize the successful clinical outcome for the patient, this review article intends to discuss the latest developments in the genetic and molecular mechanisms of cancer progression and treatment resistance, and how they influence the planning of the treatment strategies of HER2-positive breast cancers.
Collapse
Affiliation(s)
- Christopher Y.C. Chow
- UNIMED Medical Institute, Hong Kong, Hong Kong SAR, China
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | | | - Cheng-Hsun Wu
- Department of Anatomy, China Medical University, Taichung, Taiwan
| | - Louis W.C. Chow
- UNIMED Medical Institute, Hong Kong, Hong Kong SAR, China
- Organisation for Oncology and Translational Research, Hong Kong, Hong Kong SAR, China
- *Correspondence: Louis W.C. Chow,
| |
Collapse
|
17
|
Eskandarion MR, Tizmaghz Z, Andalib B, Parsa N, Emami SAH, Shahsiah R, Oghabian MA, Shirkoohi R. A case report of the sustained and rapid response of bevacizumab in a TP53-positive breast cancer and liver metastatic patient through personalized medicine. Front Oncol 2022; 12:940678. [PMID: 36119510 PMCID: PMC9479335 DOI: 10.3389/fonc.2022.940678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/08/2022] [Indexed: 11/28/2022] Open
Abstract
HER2-positive metastatic breast cancer is much less frequent than other subgroups of breast cancer. Treatment options for this cancer are mostly limited to systemic chemotherapy, which leads to moderate improvements. Targeted therapy against malignant breast cancer requires the identification of reliable biomarkers for personalized medicine to obtain the maximum benefit of this therapy. Any mutations in the TP53 signaling pathway can be considered as a significant causative factor of breast cancer, for which the identification of target genes plays an important role in selecting the appropriate treatment. The use of personalized gene expression profiling could be valuable to find the direct target of the treatment in this case. The present study assessed the genetic profile of an HER2-positive metastatic breast cancer patient (with a liver metastasis) and figured out a complete and sustained response to bevacizumab. According to the results of next-generation sequencing (NGS) analysis, the patient’s genetic profile showed an increased expression of p4EBP1 and PTEN and the activation of the mTOR signaling pathway with a mutation in the TP53 gene. Based on the common treatment of similar profiling, we administrated bevacizumab/Taxol/Gemzar chemotherapy up to six courses. Accordingly, as the response to treatment was revealed by reducing the volume of the liver metastasis from 4 to 1.4 cm, metastasectomy was performed as a complementary treatment. Hence, personalized gene expression profiling not only is useful for targeted therapy but also could be recommended to avoid prescription of non-responsive drugs.
Collapse
Affiliation(s)
- Mohammad Reza Eskandarion
- Cancer Research Center, Cancer Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- BESTforPM (Biomarker Evaluation and Supervision Team for Personalized Medicine), Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Tizmaghz
- BESTforPM (Biomarker Evaluation and Supervision Team for Personalized Medicine), Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Department of Radiation Oncology, Cancer Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahram Andalib
- BESTforPM (Biomarker Evaluation and Supervision Team for Personalized Medicine), Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Radiation Oncology Central Clinic of Karaj (ROCK), Karaj, Iran
- *Correspondence: Reza Shirkoohi, ; Bahram Andalib,
| | - Nasser Parsa
- BESTforPM (Biomarker Evaluation and Supervision Team for Personalized Medicine), Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Iranian Cancer Association, Tehran, Iran
| | - Seyed Amir Hossein Emami
- Cancer Research Center, Cancer Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- BESTforPM (Biomarker Evaluation and Supervision Team for Personalized Medicine), Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Department of Hematology and Medical Oncology, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Shahsiah
- BESTforPM (Biomarker Evaluation and Supervision Team for Personalized Medicine), Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Department of Anatomical and Clinical Pathology, Cancer Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Oghabian
- BESTforPM (Biomarker Evaluation and Supervision Team for Personalized Medicine), Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Medical Physics Department, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Shirkoohi
- Cancer Research Center, Cancer Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- BESTforPM (Biomarker Evaluation and Supervision Team for Personalized Medicine), Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- *Correspondence: Reza Shirkoohi, ; Bahram Andalib,
| |
Collapse
|
18
|
Mammari N, Salles E, Beaussart A, El-Kirat-Chatel S, Varbanov M. Squalamine and Its Aminosterol Derivatives: Overview of Biological Effects and Mechanisms of Action of Compounds with Multiple Therapeutic Applications. Microorganisms 2022; 10:microorganisms10061205. [PMID: 35744723 PMCID: PMC9229800 DOI: 10.3390/microorganisms10061205] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 05/30/2022] [Accepted: 06/07/2022] [Indexed: 02/04/2023] Open
Abstract
Squalamine is a natural aminosterol that has been discovered in the tissues of the dogfish shark (Squalus acanthias). Studies have previously demonstrated that this promoter compound and its derivatives exhibit potent bactericidal activity against Gram-negative, Gram-positive bacteria, and multidrug-resistant bacteria. The antibacterial activity of squalamine was found to correlate with that of other antibiotics, such as colistin and polymyxins. Still, in the field of microbiology, evidence has shown that squalamine and its derivatives have antifungal activity, antiprotozoa effect against a limited list of protozoa, and could exhibit antiviral activity against both RNA- and DNA-enveloped viruses. Furthermore, squalamine and its derivatives have been identified as being antiangiogenic compounds in the case of several types of cancers and induce a potential positive effect in the case of other diseases such as experimental retinopathy and Parkinson's disease. Given the diverse effects of the squalamine and its derivatives, in this review we provide the different advances in our understanding of the various effects of these promising molecules and try to draw up a non-exhaustive list of the different mechanisms of actions of squalamine and its derivatives on the human organism and on different pathogens.
Collapse
Affiliation(s)
- Nour Mammari
- Université de Lorraine, CNRS, L2CM, F-54000 Nancy, France; (N.M.); (E.S.)
| | - Elsa Salles
- Université de Lorraine, CNRS, L2CM, F-54000 Nancy, France; (N.M.); (E.S.)
| | | | | | - Mihayl Varbanov
- Université de Lorraine, CNRS, L2CM, F-54000 Nancy, France; (N.M.); (E.S.)
- Laboratoire de Virologie, CHRU de Nancy Brabois, F-54500 Vandœuvre-lès-Nancy, France
- Correspondence:
| |
Collapse
|
19
|
Stereotactic Body Radiation in Breast Cancer — Definitive, Oligometastatic, and Beyond. CURRENT BREAST CANCER REPORTS 2022. [DOI: 10.1007/s12609-022-00447-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
20
|
Role of Anti-Angiogenic Factors in the Pathogenesis of Breast Cancer: A Review of Therapeutic Potential. Pathol Res Pract 2022; 236:153956. [DOI: 10.1016/j.prp.2022.153956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/06/2022] [Accepted: 05/25/2022] [Indexed: 11/23/2022]
|
21
|
Umemneku-Chikere CM, Ayodele O, Soares M, Khan S, Abrams K, Owen R, Bujkiewicz S. Comparative review of pharmacological therapies in individuals with HER2-positive advanced breast cancer with focus on hormone receptor subgroups. Front Oncol 2022; 12:943154. [PMID: 36059633 PMCID: PMC9433866 DOI: 10.3389/fonc.2022.943154] [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: 05/13/2022] [Accepted: 07/22/2022] [Indexed: 11/13/2022] Open
Abstract
Breast cancer is the fifth leading cause of cancer-related deaths worldwide. The randomized controlled trials (RCTs) of targeted therapies in human epidermal receptor 2 (HER2)-positive advanced breast cancer (ABC) have provided an evidence base for regulatory and reimbursement agencies to appraise the use of cancer therapies in clinical practice. However, a subset of these patients harbor additional biomarkers, for example, a positive hormone receptor status that may be more amenable to therapy and improve overall survival (OS). This review seeks to explore the reporting of evidence for treatment effects by the hormone receptor status using the RCT evidence of targeted therapies for HER2-positive ABC patients. Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines were followed to identify published RCTs. Extracted data were synthesized using network meta-analysis to obtain the relative effects of HER2-positive-targeted therapies. We identified a gap in the reporting of the effectiveness of therapies by the hormone receptor status as only 15 out of 42 identified RCTs reported hormone receptor subgroup analyses; the majority of which reported progression-free survival but not OS or the overall response rate. In conclusion, we recommend that future trials in ABC should report the effect of cancer therapies in hormone receptor subgroups for all outcomes.
Collapse
Affiliation(s)
| | - Olubukola Ayodele
- University Hospital Leicester National Health Service (NHS) Trust, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Marta Soares
- Centre for Health Economics, University of York, York, United Kingdom
| | - Sam Khan
- Leicester Cancer Research Centre, University of Leicester, Leicester, United Kingdom
| | - Keith Abrams
- Department of Statistics, University of Warwick, Coventry, United Kingdom
| | - Rhiannon Owen
- Medical School, Swansea University, Swansea, United Kingdom
| | - Sylwia Bujkiewicz
- Biostatistics Research Group, Department of Health Sciences, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
22
|
Farahmand S, Fernandez AI, Ahmed FS, Rimm DL, Chuang JH, Reisenbichler E, Zarringhalam K. Deep learning trained on hematoxylin and eosin tumor region of Interest predicts HER2 status and trastuzumab treatment response in HER2+ breast cancer. Mod Pathol 2022; 35:44-51. [PMID: 34493825 DOI: 10.1038/s41379-021-00911-w] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 08/13/2021] [Accepted: 08/13/2021] [Indexed: 12/19/2022]
Abstract
The current standard of care for many patients with HER2-positive breast cancer is neoadjuvant chemotherapy in combination with anti-HER2 agents, based on HER2 amplification as detected by in situ hybridization (ISH) or protein immunohistochemistry (IHC). However, hematoxylin & eosin (H&E) tumor stains are more commonly available, and accurate prediction of HER2 status and anti-HER2 treatment response from H&E would reduce costs and increase the speed of treatment selection. Computational algorithms for H&E have been effective in predicting a variety of cancer features and clinical outcomes, including moderate success in predicting HER2 status. In this work, we present a novel convolutional neural network (CNN) approach able to predict HER2 status with increased accuracy over prior methods. We trained a CNN classifier on 188 H&E whole slide images (WSIs) manually annotated for tumor Regions of interest (ROIs) by our pathology team. Our classifier achieved an area under the curve (AUC) of 0.90 in cross-validation of slide-level HER2 status and 0.81 on an independent TCGA test set. Within slides, we observed strong agreement between pathologist annotated ROIs and blinded computational predictions of tumor regions / HER2 status. Moreover, we trained our classifier on pre-treatment samples from 187 HER2+ patients that subsequently received trastuzumab therapy. Our classifier achieved an AUC of 0.80 in a five-fold cross validation. Our work provides an H&E-based algorithm that can predict HER2 status and trastuzumab response in breast cancer at an accuracy that may benefit clinical evaluations.
Collapse
Affiliation(s)
- Saman Farahmand
- University of Massachusetts-Boston, Department of Mathematics, Boston, MA, USA.,University of Massachusetts-Boston, Computational Sciences PhD Program, Boston, MA, USA
| | - Aileen I Fernandez
- Yale University, Yale School of Medicine, Department of Pathology, New Haven, CT, USA
| | - Fahad Shabbir Ahmed
- Yale University, Yale School of Medicine, Department of Pathology, New Haven, CT, USA
| | - David L Rimm
- Yale University, Yale School of Medicine, Department of Pathology, New Haven, CT, USA
| | - Jeffrey H Chuang
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA. .,UCONN Health, Department of Genetics and Genome Sciences, Farmington, CT, USA.
| | - Emily Reisenbichler
- Yale University, Yale School of Medicine, Department of Pathology, New Haven, CT, USA.
| | - Kourosh Zarringhalam
- University of Massachusetts-Boston, Department of Mathematics, Boston, MA, USA. .,University of Massachusetts-Boston, Computational Sciences PhD Program, Boston, MA, USA.
| |
Collapse
|
23
|
Cardoso F, Cella D, Velikova G, Harmer V, Schumacher-Wulf E, Rihani J, Casas A, Harbeck N. Quality-of-life methodology in hormone receptor-positive advanced breast cancer: Current tools and perspectives for the future. Cancer Treat Rev 2021; 102:102321. [PMID: 34852292 DOI: 10.1016/j.ctrv.2021.102321] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 11/10/2021] [Accepted: 11/15/2021] [Indexed: 12/20/2022]
Abstract
Health-related quality of life (HRQOL) is increasingly recognized as important when evaluating cancer treatments. The use, reporting, and analysis of patient-reported outcome measures (PROMs), however, are not standardized in clinical trials and are often poorly implemented in clinical practice. We report the results of a systematic literature review (PubMed search: January 1, 2000 to August 15, 2020) of PROM use, reporting, and analysis in phase 3 clinical trials of hormone receptor-positive (HR+) advanced breast cancer (ABC). Further inspection of cyclin-dependent kinase 4/6 (CDK4/6) inhibitor publications was performed to examine PROMs in the HR+/human epidermal growth factor receptor 2-negative setting. A total of 88 results were identified in the initial search; 32 were included in the final analysis. Among included studies, most (66%) had been published in the last 5 years (2015 to 2020). CDK4/6 inhibitors (38%) were the most common agents reported. No clear standard for PROM use, reporting, or analysis was found. The most common PROMs were European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire Core 30 (EORTC QLQ-C30; 59%) and the Functional Assessment of Cancer Therapy-Breast (FACT-B; 34%). Important differences, among studies that reported them, ranged from 5 to 10 points for the EORTC QLQ-C30 and 8 points for the FACT-B total score. This review showed that a lack of clear consistency remains for PROM use, reporting, and analysis in phase 3 clinical trials of HR+ ABC. However, HRQOL is of high interest in the literature, including for CDK4/6 inhibitors.
Collapse
Affiliation(s)
- Fatima Cardoso
- Breast Unit, Champalimaud Clinical Center/Champalimaud Foundation, Lisbon, Portugal.
| | - David Cella
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Galina Velikova
- Leeds Institute of Medical Research at St. James's University of Leeds and Leeds Teaching Hospitals, Leeds, UK
| | | | | | | | - Ana Casas
- University Hospital Virgen del Rocio (HUVR), Sevilla, Spain
| | - Nadia Harbeck
- Breast Center, Department of Obstetrics and Gynecology, LMU University Hospital Munich, Germany
| |
Collapse
|
24
|
Mansouri A, Padmanaban V, Aregawi D, Glantz M. VEGF and Immune Checkpoint Inhibition for Prevention of Brain Metastases: Systematic Review and Meta-analysis. Neurology 2021; 97:e1484-e1492. [PMID: 34380750 DOI: 10.1212/wnl.0000000000012642] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 07/14/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES We conducted a systematic review and meta-analysis to investigate the role of vascular endothelial growth factor (VEGF) inhibitors and immune checkpoint inhibitors (ICIs) in preventing the development of brain metastases (BMs). METHODS We searched MEDLINE, Embase, Cochrane Database, and Google Scholar between January 1, 2000, and June 1, 2020. Included studies were randomized controlled trials (RCTs) of adults with systemic cancer that reported the incidence of BMs treated with and without VEGF inhibitors, as well as observational studies of adults with systemic cancer that reported the incidence of BMs treated with and without ICIs (no RCTs addressed the ICI question). Pooled relative risks (RR) were computed with a binary random-effects model. RESULTS A search for VEGF and incidence of new BMs revealed 7 studies (6,212 patients with breast, colon, and non-small-cell lung cancer). Meta-analysis showed a lower incidence of new BMs compared to control (RR 0.71, 95% confidence interval [CI] 0.56-0.89, p = 0.003). A search for ICIs and incidence of new BMs yielded 8 studies (732 patients with non-small-cell lung cancer or metastatic melanoma) in which ICIs were used as an adjunct to radiosurgery. Meta-analysis showed a lower incidence of out-of-treatment-field BMs with ICIs compared to controls at 1 year (RR 0.65, 95% CI 0.49-0.88, p = 0.005). The overall Grading of Recommendations, Assessment, Development and Evaluations score for the evidence evaluating the role of bevacizumab and ICIs was high and moderate, respectively. DISCUSSION VEGF and ICIs may have a role in prophylaxis against BM in patients with solid tumors.
Collapse
Affiliation(s)
- Alireza Mansouri
- From the Departments of Neurosurgery (A.M., V.P., D.A., M.G.), Neurology (D.A., M.G.), and Oncology (D.A., M.G.), Penn State Milton S. Hershey Medical Center, Hershey, PA
| | - Varun Padmanaban
- From the Departments of Neurosurgery (A.M., V.P., D.A., M.G.), Neurology (D.A., M.G.), and Oncology (D.A., M.G.), Penn State Milton S. Hershey Medical Center, Hershey, PA
| | - Dawit Aregawi
- From the Departments of Neurosurgery (A.M., V.P., D.A., M.G.), Neurology (D.A., M.G.), and Oncology (D.A., M.G.), Penn State Milton S. Hershey Medical Center, Hershey, PA
| | - Michael Glantz
- From the Departments of Neurosurgery (A.M., V.P., D.A., M.G.), Neurology (D.A., M.G.), and Oncology (D.A., M.G.), Penn State Milton S. Hershey Medical Center, Hershey, PA.
| |
Collapse
|
25
|
Hamada-Nishimoto M, Kang Y, Shiraki E, Tsuyuki S. Potential effectiveness of combining bevacizumab with paclitaxel for treating HER2-positive metastatic breast cancer. Int Cancer Conf J 2021; 10:329-333. [PMID: 34567947 DOI: 10.1007/s13691-021-00504-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 07/23/2021] [Indexed: 11/25/2022] Open
Abstract
Although various first- to third-line HER2-targeted therapies have been established, the optimal sequence after third-line therapy has not been determined yet. Here, we describe seven patients with HER2-positive metastatic breast cancer who underwent bevacizumab (BV) and paclitaxel (PTX) combination therapy after several HER2-targeted therapies. Re-biopsy of metastatic sites was performed on four patients during the treatment prior to BV + PTX; three patients presented the same HER2-positive status as that of the primary tumor and two of whom achieved partial response. Four of seven patients achieved partial response and a 10-month median progression-free survival. Therefore, bevacizumab combined with paclitaxel is potentially effective in the treatment of HER2-positive metastatic breast cancer, if standard HER2-targeted therapy fails.
Collapse
Affiliation(s)
- Mai Hamada-Nishimoto
- Department of Breast Surgery, Osaka Red Cross Hospital, 5-30 Fudegasaki-cho, Tennoji-ku, Osaka, 543-8555 Japan
| | - Yookija Kang
- Department of Breast Surgery, Osaka Red Cross Hospital, 5-30 Fudegasaki-cho, Tennoji-ku, Osaka, 543-8555 Japan
| | - Eriko Shiraki
- Department of Breast Surgery, Osaka Red Cross Hospital, 5-30 Fudegasaki-cho, Tennoji-ku, Osaka, 543-8555 Japan
| | - Shigeru Tsuyuki
- Department of Breast Surgery, Osaka Red Cross Hospital, 5-30 Fudegasaki-cho, Tennoji-ku, Osaka, 543-8555 Japan
| |
Collapse
|
26
|
Larbouret C, Gros L, Pèlegrin A, Chardès T. Improving Biologics' Effectiveness in Clinical Oncology: From the Combination of Two Monoclonal Antibodies to Oligoclonal Antibody Mixtures. Cancers (Basel) 2021; 13:cancers13184620. [PMID: 34572847 PMCID: PMC8465647 DOI: 10.3390/cancers13184620] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/01/2021] [Accepted: 09/09/2021] [Indexed: 01/03/2023] Open
Abstract
Simple Summary The approval of the two antibody combinations trastuzumab/pertuzumab and ipilimumab/nivolumab in oncology has paved the way for novel antibody combinations or oligoclonal antibody mixtures to improve their efficacy in cancer. The underlying biological mechanisms and challenges of these strategies will be discussed using data from clinical trials listed in databases. These therapeutic combinations also lead to questions on how to optimize their formulation and delivery to induce a therapeutic polyclonal response in patients with cancer. Abstract Monoclonal antibodies have revolutionized the treatment of many diseases, but their clinical efficacy remains limited in some other cases. Pre-clinical and clinical trials have shown that combinations of antibodies that bind to the same target (homo-combinations) or to different targets (hetero-combinations) to mimic the polyclonal humoral immune response improve their therapeutic effects in cancer. The approval of the trastuzumab/pertuzumab combination for breast cancer and then of the ipilimumab/nivolumab combination for melanoma opened the way to novel antibody combinations or oligoclonal antibody mixtures as more effective biologics for cancer management. We found more than 300 phase II/III clinical trials on antibody combinations, with/without chemotherapy, radiotherapy, small molecules or vaccines, in the ClinicalTrials.gov database. Such combinations enhance the biological responses and bypass the resistance mechanisms observed with antibody monotherapy. Usually, such antibody combinations are administered sequentially as separate formulations. Combined formulations have also been developed in which separately produced antibodies are mixed before administration or are produced simultaneously in a single cell line or a single batch of different cell lines as a polyclonal master cell bank. The regulation, toxicity and injection sequence of these oligoclonal antibody mixtures still need to be addressed in order to optimize their delivery and their therapeutic effects.
Collapse
Affiliation(s)
- Christel Larbouret
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Institut Régional du Cancer de Montpellier (ICM), Inserm U1194, Université de Montpellier, 34298 Montpellier, France; (L.G.); (A.P.); (T.C.)
- Correspondence: ; Tel.: +33-411-283-110
| | - Laurent Gros
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Institut Régional du Cancer de Montpellier (ICM), Inserm U1194, Université de Montpellier, 34298 Montpellier, France; (L.G.); (A.P.); (T.C.)
- Centre National de la Recherche Scientifique (CNRS), 75016 Paris, France
| | - André Pèlegrin
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Institut Régional du Cancer de Montpellier (ICM), Inserm U1194, Université de Montpellier, 34298 Montpellier, France; (L.G.); (A.P.); (T.C.)
| | - Thierry Chardès
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Institut Régional du Cancer de Montpellier (ICM), Inserm U1194, Université de Montpellier, 34298 Montpellier, France; (L.G.); (A.P.); (T.C.)
- Centre National de la Recherche Scientifique (CNRS), 75016 Paris, France
| |
Collapse
|
27
|
Zhang X, Leng J, Zhou Y, Mao F, Lin Y, Shen S, Sun Q. Efficacy and Safety of Anti-HER2 Agents in Combination With Chemotherapy for Metastatic HER2-Positive Breast Cancer Patient: A Network Meta-Analysis. Front Oncol 2021; 11:731210. [PMID: 34490125 PMCID: PMC8416996 DOI: 10.3389/fonc.2021.731210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 07/27/2021] [Indexed: 01/01/2023] Open
Abstract
Background The presence of anti-HER2 agents, such as trastuzumab, pertuzumab, and trastuzumab emtansine (T-DM1), significantly improved the prognosis of metastatic HER2-positive (HER2+) breast cancers (BC). However, drug resistance and disease progression are still common. In order to further improve the treatment efficacy, new clinical trials about anti-HER2 agents in combination with chemotherapy are growing rapidly. We conducted the network meta-analysis to synthesize evidences of clinical trials to identify the best therapy for metastatic HER2+ BC. Methods A systematic search of randomized controlled trials regarding anti-HER2 agents in combination with chemotherapy for advanced or metastatic breast cancers up to May 2020 was conducted in Embase, PubMed, and the Cochrane Library. The primary outcome was progression-free survival (PFS). The secondary outcomes were overall survival (OS), objective response rate (ORR), and safety. Bayesian network meta-analysis was conducted to synthesize the results and rank the therapies. Results Twenty-six studies, including 16 studies for first-line treatments and 10 studies for second- or later-line treatments were included in the network meta-analysis. For first-line studies, the THP (taxanes + trastuzumab + pertuzumab) regimen exhibited the highest probability to be the optimal treatment in all efficacy outcomes and moderate safety. For second- or later-line studies, the T-DM1 and XHTuC (capecitabine + trastuzumab + tucatinib) regimens ranked top two in all efficacy outcomes according to the surface under the cumulative ranking (SUCRA) results. T-DM1 ranked first in PFS and OS whereas XHTuC ranked first in ORR. The safety outcomes of T-DM1 and XHTuC were acceptable. Conclusions THP was still the optimal first-line treatment for metastatic HER2+ BC. T-DM1 and XHTuC were recommended for second-line treatments. Systematic Review Registration INPLASY.com, identifier (INPLASY202090086).
Collapse
Affiliation(s)
- Xiaohui Zhang
- Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Junsheng Leng
- Department of Orthopedics, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Yidong Zhou
- Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Feng Mao
- Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Yan Lin
- Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Songjie Shen
- Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences (CAMS), Beijing, China
| | - Qiang Sun
- Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences (CAMS), Beijing, China
| |
Collapse
|
28
|
de Melo Bisneto AV, Fernandes AS, Velozo Sá VDS, Véras JH, Soares ETS, da Silva Santos AF, Cardoso CG, Silveira-Lacerda EDP, Carneiro CC, Chen-Chen L. Anti-angiogenic activity of azathioprine. Microvasc Res 2021; 138:104234. [PMID: 34478745 DOI: 10.1016/j.mvr.2021.104234] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/16/2021] [Accepted: 08/20/2021] [Indexed: 11/16/2022]
Abstract
Azathioprine (AZA) is the main drug used in immunomodulatory therapy in post-transplant patients or with autoimmune diseases. However, no study has evaluated the AZA angiogenic response. Therefore, this study investigated the effects of AZA on the angiogenic process through macroscopic, histological, and immunohistochemical analyses in chick embryo chorioallantoic membrane (CAM). Our results showed potent anti-angiogenic activity of AZA at the higher concentrations tested in the CAM assay. The histological analysis of CAM confirmed this effect, since AZA induced a significant reduction in all parameters evaluated. In addition, immunohistochemical evaluation of CAM revealed that AZA decreased TGF-β and VEGF levels, important cytokines involved in the angiogenic process. Therefore, the AZA anti-angiogenic effect identified in our study provides new information for the possible application of this drug in anticancer treatment.
Collapse
Affiliation(s)
- Abel Vieira de Melo Bisneto
- Laboratory of Radiobiology and Mutagenesis, Department of Genetics of Institute of Biological Sciences, Federal University of Goiás, 74690-900 Goiânia, Brazil
| | - Amanda Silva Fernandes
- Laboratory of Radiobiology and Mutagenesis, Department of Genetics of Institute of Biological Sciences, Federal University of Goiás, 74690-900 Goiânia, Brazil
| | - Vivianne de Souza Velozo Sá
- Laboratory of Molecular Genetics and Cytogenetics, Department of Genetics of Institute of Biological Sciences, Federal University of Goiás, 74690-900 Goiânia, Brazil
| | - Jefferson Hollanda Véras
- Laboratory of Radiobiology and Mutagenesis, Department of Genetics of Institute of Biological Sciences, Federal University of Goiás, 74690-900 Goiânia, Brazil
| | - Estéfane Thaíne Sodré Soares
- Laboratory of Radiobiology and Mutagenesis, Department of Genetics of Institute of Biological Sciences, Federal University of Goiás, 74690-900 Goiânia, Brazil
| | | | - Clever Gomes Cardoso
- Department of Morphology of Institute of Biological Sciences, Federal University of Goiás, 74690-900 Goiânia, Brazil
| | - Elisângela de Paula Silveira-Lacerda
- Laboratory of Molecular Genetics and Cytogenetics, Department of Genetics of Institute of Biological Sciences, Federal University of Goiás, 74690-900 Goiânia, Brazil
| | - Cristiene Costa Carneiro
- Institute of Health Sciences, Universidade Paulista - Campus Flamboyant, 74845-090 Goiânia, Brazil
| | - Lee Chen-Chen
- Laboratory of Radiobiology and Mutagenesis, Department of Genetics of Institute of Biological Sciences, Federal University of Goiás, 74690-900 Goiânia, Brazil.
| |
Collapse
|
29
|
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
|
30
|
Ribatti D, Solimando AG, Pezzella F. The Anti-VEGF(R) Drug Discovery Legacy: Improving Attrition Rates by Breaking the Vicious Cycle of Angiogenesis in Cancer. Cancers (Basel) 2021; 13:cancers13143433. [PMID: 34298648 PMCID: PMC8304542 DOI: 10.3390/cancers13143433] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 06/24/2021] [Accepted: 07/06/2021] [Indexed: 02/07/2023] Open
Abstract
Resistance to anti-vascular endothelial growth factor (VEGF) molecules causes lack of response and disease recurrence. Acquired resistance develops as a result of genetic/epigenetic changes conferring to the cancer cells a drug resistant phenotype. In addition to tumor cells, tumor endothelial cells also undergo epigenetic modifications involved in resistance to anti-angiogenic therapies. The association of multiple anti-angiogenic molecules or a combination of anti-angiogenic drugs with other treatment regimens have been indicated as alternative therapeutic strategies to overcome resistance to anti-angiogenic therapies. Alternative mechanisms of tumor vasculature, including intussusceptive microvascular growth (IMG), vasculogenic mimicry, and vascular co-option, are involved in resistance to anti-angiogenic therapies. The crosstalk between angiogenesis and immune cells explains the efficacy of combining anti-angiogenic drugs with immune check-point inhibitors. Collectively, in order to increase clinical benefits and overcome resistance to anti-angiogenesis therapies, pan-omics profiling is key.
Collapse
Affiliation(s)
- Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, 70124 Bari, Italy
- Correspondence: ; Tel.: +39-080-547832
| | - Antonio Giovanni Solimando
- Guido Baccelli Unit of Internal Medicine, Department of Biomedical Sciences and Human Oncology, School of Medicine, Aldo Moro University of Bari, 70124 Bari, Italy;
- IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy
| | - Francesco Pezzella
- Nuffield Division of Laboratory Science, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX39DU, UK;
| |
Collapse
|
31
|
Anti-HER2 antibody prolongs overall survival disproportionally more than progression-free survival in HER2-Positive metastatic breast cancer patients. Breast 2021; 59:211-220. [PMID: 34298300 PMCID: PMC8321956 DOI: 10.1016/j.breast.2021.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/28/2021] [Accepted: 07/05/2021] [Indexed: 12/01/2022] Open
Abstract
Background This meta-analysis aimed to test the hypothesis that the HER2-positive metastatic breast cancer (mBC) patients treated with anti-HER2 antibodies in trial intervention arms have a greater prolongation of overall survival (OS) than of progression-free survival (PFS) and this extra-prolongation of median survival time in OS relates specifically to the anti-HER2 antibody. Methods The NCBI/Pubmed and Cochrane databases were searched systematically for HER2-positive or mBC trials published in English during January 1999–November 2017. Treatment arms with shorter PFS were considered as the “control” arm, whereas those with longer PFS as the “test” arm. The between-treatment drug differences were grouped into nine categories. Groups with or without anti-HER2 antibodies were pooled respectively for comparisons. The interrelationships between PFS and OS hazard ratios (HRs) and median survival time differences were investigated by conducting fixed-effects and mixed-effects linear meta-regression analyses. Results Twenty-eight trials (10,928 patients) from 438 articles were collected, and four with missing data were excluded in meta-regression analysis. Overall median PFS (HR = 0.73, 95% CI: 0.68–0.78) and median OS (HR = 0.82, 95% CI: 0.77–0.87) weakly favored the longer PFS arm with a weak correlation between the PFS and OS HRs. However, the between-treatment drug difference was anti-HER2 antibody, the absolute increment in median OS time was double that of median PFS time (p < 0.001) and linearly correlated, which was not found with any non-anti-HER2 antibody drug differences. Conclusions Anti-HER2 antibody in patients with HER2-positive mBC prolonged OS more than PFS and mandates further investigation. Our study dissected the overall survival benefit over progression-free survival in HER-2 positive metastatic breast cancer treated with anti-HER2 antibody by meta-analysis. Use of anti-HER2 antibodies, but not other anticancer drugs, to treat HER2-positive metastatic breast cancer independently predicted the OS compared with PFS. The design of future clinical trials should consider anti-HER2 antibodies carefully in the treatment armamentarium for HER2-positive metastatic breast cancer.
Collapse
|
32
|
Yin G, Zhao L. Risk of hypertension with anti-VEGF monoclonal antibodies in cancer patients: a systematic review and meta-analysis of 105 phase II/III randomized controlled trials. J Chemother 2021; 34:221-234. [PMID: 34229563 DOI: 10.1080/1120009x.2021.1947022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
We performed a meta-analysis to fully investigate the hypertension of anti-VEGF mAbs in cancer patients. Databases were searched for randomized controlled trials (RCTs) treated with anti-VEGF mAbs till January 2021. The relevant RCTs in cancer patients treated with anti-VEGF mAbs were retrieved and the systematic evaluation was conducted. One hundred and five RCTs and 65358 patients were included. Our study suggests that anti-VEGF mAbs significantly increased the risks of all-grade (RR, 3.22; 95%CI, 2.83-3.65; p < 0.00001; I2=71%) and high-grade (RR, 6.15; 95%CI, 5.58-6.78; p < 0.00001; I2=48%) hypertension in cancer patients. Those risks may be dependent on drug type. Icrucumab did not association with an increased risk of hypertension. The RR of hypertension did not vary significantly according to the type of cancer, line of therapy, and treatment duration. The available data suggested that the use of anti-VEGF mAbs were associated with a significantly increased risk of hypertension.
Collapse
Affiliation(s)
- Gang Yin
- Central Nervous System Drug Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou, Sichuan, P.R. China.,Engineering Research Center for Pharmaceuticals and Equipments of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of pharmacy, Chengdu University, Chengdu, Sichuan, P.R. China
| | - Ling Zhao
- Central Nervous System Drug Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou, Sichuan, P.R. China
| |
Collapse
|
33
|
Chainitikun S, Saleem S, Lim B, Valero V, Ueno NT. Update on systemic treatment for newly diagnosed inflammatory breast cancer. J Adv Res 2021; 29:1-12. [PMID: 33842000 PMCID: PMC8020152 DOI: 10.1016/j.jare.2020.08.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 07/31/2020] [Accepted: 08/21/2020] [Indexed: 12/14/2022] Open
Abstract
Background Inflammatory breast cancer (IBC) is a rare and aggressive disease, accounting for 2-4% of new cases of breast cancer. Owing to its aggressive nature, IBC represent approximately 8-10% of breast cancer deaths. Management of IBC requires a multidisciplinary team for decision-making involving a composite of systemic treatment, surgery, and radiation, or "Trimodality Treatment." Because of the rarity of the disease, systemic therapy of IBC traditionally has been extrapolated from non-IBC clinical trials. Aim of Review The purpose of this review is to provide an overview of the development of systemic treatment of IBC from the past to the present by focusing on IBC clinical trials, including chemotherapy and targeted therapies. Key Scientific Concepts of Review We discuss their effects on pathologic complete response (pCR) and survival outcomes, the predictive markers, and the adverse events of these therapies. Further, we summarized the current standard treatment stratified by molecular subtypes based on clinical data. Finally, we discuss the future trend of systemic therapy, including immunotherapy and ongoing IBC clinical trials.
Collapse
Affiliation(s)
- Sudpreeda Chainitikun
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Sadia Saleem
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Bora Lim
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Vicente Valero
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Naoto T. Ueno
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
34
|
Vacchelli E, Aranda F, Eggermont A, Galon J, Sautès-Fridman C, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Tumor-targeting monoclonal antibodies in cancer therapy. Oncoimmunology 2021; 3:e27048. [PMID: 24605265 PMCID: PMC3937194 DOI: 10.4161/onci.27048] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 11/01/2013] [Indexed: 02/06/2023] Open
Abstract
In 1997, for the first time in history, a monoclonal antibody (mAb), i.e., the chimeric anti-CD20 molecule rituximab, was approved by the US Food and Drug Administration for use in cancer patients. Since then, the panel of mAbs that are approved by international regulatory agencies for the treatment of hematopoietic and solid malignancies has not stopped to expand, nowadays encompassing a stunning amount of 15 distinct molecules. This therapeutic armamentarium includes mAbs that target tumor-associated antigens, as well as molecules that interfere with tumor-stroma interactions or exert direct immunostimulatory effects. These three classes of mAbs exert antineoplastic activity via distinct mechanisms, which may or may not involve immune effectors other than the mAbs themselves. In previous issues of OncoImmunology, we provided a brief scientific background to the use of mAbs, all types confounded, in cancer therapy, and discussed the results of recent clinical trials investigating the safety and efficacy of this approach. Here, we focus on mAbs that primarily target malignant cells or their interactions with stromal components, as opposed to mAbs that mediate antineoplastic effects by activating the immune system. In particular, we discuss relevant clinical findings that have been published during the last 13 months as well as clinical trials that have been launched in the same period to investigate the therapeutic profile of hitherto investigational tumor-targeting mAbs.
Collapse
Affiliation(s)
- Erika Vacchelli
- Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France ; Université Paris-Sud/Paris XI; Paris, France
| | - Fernando Aranda
- Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France
| | | | - Jérôme Galon
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France ; Université Pierre et Marie Curie/Paris VI; Paris, France ; INSERM, U872; Paris, France ; Equipe 15, Centre de Recherche des Cordeliers; Paris, France
| | - Catherine Sautès-Fridman
- Université Pierre et Marie Curie/Paris VI; Paris, France ; INSERM, U872; Paris, France ; Equipe 13, Centre de Recherche des Cordeliers; Paris, France
| | - Laurence Zitvogel
- Gustave Roussy; Villejuif, France ; INSERM, U1015; CICBT507; Villejuif, France
| | - Guido Kroemer
- Pôle de Biologie; Hôpital Européen Georges Pompidou; AP-HP; Paris, France ; Metabolomics and Cell Biology Platforms; Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| | - Lorenzo Galluzzi
- Gustave Roussy; Villejuif, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France
| |
Collapse
|
35
|
Leone JP, Emblem KE, Weitz M, Gelman RS, Schneider BP, Freedman RA, Younger J, Pinho MC, Sorensen AG, Gerstner ER, Harris G, Krop IE, Morganstern D, Sohl J, Hu J, Kasparian E, Winer EP, Lin NU. Phase II trial of carboplatin and bevacizumab in patients with breast cancer brain metastases. Breast Cancer Res 2020; 22:131. [PMID: 33256829 PMCID: PMC7706261 DOI: 10.1186/s13058-020-01372-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 11/15/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND We aimed to examine the safety and efficacy of bevacizumab and carboplatin in patients with breast cancer brain metastases. METHODS We enrolled patients with breast cancer and > 1 measurable new or progressive brain metastasis. Patients received bevacizumab 15 mg/kg intravenously (IV) on cycle 1 day 1 and carboplatin IV AUC = 5 on cycle 1 day 8. Patients with HER2-positive disease also received trastuzumab. In subsequent cycles, all drugs were administered on day 1 of each cycle. Contrast-enhanced brain MRI was performed at baseline, 24-96 h after the first bevacizumab dose (day + 1), and every 2 cycles. The primary endpoint was objective response rate in the central nervous system (CNS ORR) by composite criteria. Associations between germline VEGF single nucleotide polymorphisms (rs699947, rs2019063, rs1570360, rs833061) and progression-free survival (PFS) and overall survival (OS) were explored, as were associations between early (day + 1) MRI changes and outcomes. RESULTS Thirty-eight patients were enrolled (29 HER2-positive, 9 HER2-negative); all were evaluable for response. The CNS ORR was 63% (95% CI, 46-78). Median PFS was 5.62 months and median OS was 14.10 months. As compared with an Eastern Cooperative Oncology Group performance status (ECOG PS) of 0, patients with ECOG PS 1-2 had significantly worse PFS and OS (all P < 0.01). No significant associations between VEGF genotypes or early MRI changes and clinical outcomes were observed. CONCLUSIONS The combination of bevacizumab and carboplatin results in a high rate of durable objective response in patients with brain metastases from breast cancer. This regimen warrants further investigation. TRIAL REGISTRATION NCT01004172 . Registered 28 October 2009.
Collapse
Affiliation(s)
- Jose Pablo Leone
- Dana-Farber Cancer Institute, Dana-Farber/Brigham & Women's Cancer Center, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Kyrre E Emblem
- Department of Diagnostic Physics, Oslo University Hospital, Oslo, Norway
| | - Michelle Weitz
- Dana-Farber Cancer Institute, Dana-Farber/Brigham & Women's Cancer Center, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Rebecca S Gelman
- Dana-Farber Cancer Institute, Dana-Farber/Brigham & Women's Cancer Center, 450 Brookline Avenue, Boston, MA, 02215, USA
| | | | - Rachel A Freedman
- Dana-Farber Cancer Institute, Dana-Farber/Brigham & Women's Cancer Center, 450 Brookline Avenue, Boston, MA, 02215, USA
| | | | - Marco C Pinho
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | | | | | - Ian E Krop
- Dana-Farber Cancer Institute, Dana-Farber/Brigham & Women's Cancer Center, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Daniel Morganstern
- Dana-Farber Cancer Institute, Dana-Farber/Brigham & Women's Cancer Center, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Jessica Sohl
- Dana-Farber Cancer Institute, Dana-Farber/Brigham & Women's Cancer Center, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Jiani Hu
- Dana-Farber Cancer Institute, Dana-Farber/Brigham & Women's Cancer Center, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Elizabeth Kasparian
- Dana-Farber Cancer Institute, Dana-Farber/Brigham & Women's Cancer Center, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Eric P Winer
- Dana-Farber Cancer Institute, Dana-Farber/Brigham & Women's Cancer Center, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Nancy U Lin
- Dana-Farber Cancer Institute, Dana-Farber/Brigham & Women's Cancer Center, 450 Brookline Avenue, Boston, MA, 02215, USA.
| |
Collapse
|
36
|
Thanopoulou E, Khader L, Caira M, Wardley A, Ettl J, Miglietta F, Neven P, Guarneri V. Therapeutic Strategies for the Management of Hormone Receptor-Positive, Human Epidermal Growth Factor Receptor 2-Positive (HR+/HER2+) Breast Cancer: A Review of the Current Literature. Cancers (Basel) 2020; 12:E3317. [PMID: 33182657 PMCID: PMC7696181 DOI: 10.3390/cancers12113317] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/22/2020] [Accepted: 11/05/2020] [Indexed: 02/07/2023] Open
Abstract
Enormous advances have been made in the understanding and treatment of human epidermal growth factor receptor 2-positive breast cancer (HER2+ BC) in the last 30 years that have resulted in survival gains for affected patients. A growing body of evidence suggests that hormone receptor-positive (HR+)/HER2+ BC and HR-negative (HR-)/HER2+ BC are biologically different, with complex molecular bidirectional crosstalk between the estrogen receptor and HER2 pathway potentially affecting sensitivity to both HER2-targeted and endocrine therapy in patients with HR+/HER2+ BC. Subgroup analyses from trials enrolling patients with HER2+ BC and the results of clinical trials specifically designed to evaluate therapy in patients with HR+/HER2+ BC are helping to guide treatment decisions. In this context, encouraging results with strategies aimed at delaying or reversing drug resistance, including extended adjuvant therapy and the addition of drugs targeting alternative pathways, such as cyclin-dependent kinase (CDK) 4 and 6 inhibitors, have recently emerged. We have reached the point where tailoring the treatment according to risk and biology has become the paradigm in early BC. However, further clinical trials are needed that integrate translational research principles and identify and consider specific patient subgroups and biomarkers.
Collapse
Affiliation(s)
- Eirini Thanopoulou
- Eli Lilly and Company Limited, Erl Wood Manor, Windlesham, Surrey GU20 6PH, UK;
| | - Leila Khader
- Eli Lilly Italia S.p.A., 50019 Comune di Sesto Fiorentino, Florence, Italy; (L.K.); (M.C.)
| | - Morena Caira
- Eli Lilly Italia S.p.A., 50019 Comune di Sesto Fiorentino, Florence, Italy; (L.K.); (M.C.)
| | - Andrew Wardley
- The NIHR Manchester Clinical Research Facility at The Christie NHS Foundation Trust, School of Medical Sciences, Faculty of Biology Medicine & Health, University of Manchester, Manchester M204BX, UK;
| | - Johannes Ettl
- Department of Obstetrics and Gynecology, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany;
| | - Federica Miglietta
- Medical Oncology 2, Istituto Oncologico Veneto IOV IRCCS, 35128 Padua, Italy;
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padua, Italy
| | - Patrick Neven
- Multidisciplinary Breast Center and Department of Gynecology and Obstetrics, UZ Leuven, 3000 Leuven, Belgium;
- Department of Oncology, KU Leuven, 3000 Leuven, Belgium
| | - Valentina Guarneri
- Medical Oncology 2, Istituto Oncologico Veneto IOV IRCCS, 35128 Padua, Italy;
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padua, Italy
| |
Collapse
|
37
|
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
|
38
|
Chandra A, Rick J, Yagnik G, Aghi MK. Autophagy as a mechanism for anti-angiogenic therapy resistance. Semin Cancer Biol 2020; 66:75-88. [PMID: 31472232 PMCID: PMC7047534 DOI: 10.1016/j.semcancer.2019.08.031] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 08/27/2019] [Indexed: 02/07/2023]
Abstract
Autophagy is a lysosomal-dependent degradation process that is highly conserved and maintains cellular homeostasis by sequestering cytosolic material for degradation either non-specifically by non-selective autophagy, or targeting specific proteins aggregates by selective autophagy. Autophagy serves as a protective mechanism defending the cell from stressors and also plays an important role in enabling tumor cells to overcome harsh conditions arising in their microenvironment during growth as well as oxidative and non-oxidative injuries secondary to therapeutic stressors. Recently, autophagy has been implicated to cause tumor resistance to anti-angiogenic therapy, joining an existing literature implicating autophagy in cancer resistance to conventional DNA damaging chemotherapy and ionizing radiation. In this review, we discuss the role of angiogenesis in malignancy, mechanisms of resistance to anti-angiogenic therapy in general, the role of autophagy in driving malignancy, and the current literature in autophagy-mediated anti-angiogenic therapy resistance. Finally, we provide future insight into the current challenges of using autophagy inhibitors in the clinic and provides tips for future studies to focus on to effectively target autophagy in overcoming resistance to anti-angiogenic therapy.
Collapse
Affiliation(s)
- Ankush Chandra
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, CA, United States of America (USA); School of Medicine, Wayne State University, Detroit, MI, United States of America (USA).
| | - Jonathan Rick
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, CA, United States of America (USA).
| | - Garima Yagnik
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, CA, United States of America (USA).
| | - Manish K Aghi
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, CA, United States of America (USA).
| |
Collapse
|
39
|
Abstract
OPINION STATEMENT The management of patients with HER2+ breast cancer has evolved significantly over the preceding decades. HER2 targeting strategies have advanced beyond focusing on the receptor alone to encompass a range of approaches. Current standard of care practices in these patients relies upon dual HER2 blockade with trastuzumab and pertuzumab in the adjuvant and metastatic settings. T-DM1 has proven particularly efficacious in patients with residual disease status post neoadjuvant therapy, with additional therapies approved in the subsequent lines to address recurrent and resistant disease. Advances continue to be made in HER2+ breast cancer with multiple novel agents on the horizon, employing diverse mechanisms of action that are described in this review.
Collapse
|
40
|
Madu CO, Wang S, Madu CO, Lu Y. Angiogenesis in Breast Cancer Progression, Diagnosis, and Treatment. J Cancer 2020; 11:4474-4494. [PMID: 32489466 PMCID: PMC7255381 DOI: 10.7150/jca.44313] [Citation(s) in RCA: 142] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 04/04/2020] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis is a significant event in a wide range of healthy and diseased conditions. This process frequently involves vasodilation and an increase in vascular permeability. Numerous players referred to as angiogenic factors, work in tandem to facilitate the outgrowth of endothelial cells (EC) and the consequent vascularity. Conversely, angiogenic factors could also feature in pathological conditions. Angiogenesis is a critical factor in the development of tumors and metastases in numerous cancers. An increased level of angiogenesis is associated with decreased survival in breast cancer patients. Therefore, a good understanding of the angiogenic mechanism holds a promise of providing effective treatments for breast cancer progression, thereby enhancing patients' survival. Disrupting the initiation and progression of this process by targeting angiogenic factors such as vascular endothelial growth factor (Vegf)-one of the most potent member of the VEGF family- or by targeting transcription factors, such as Hypoxia-Inducible Factors (HIFs) that act as angiogenic regulators, have been considered potential treatment options for several types of cancers. The objective of this review is to highlight the mechanism of angiogenesis in diseases, specifically its role in the progression of malignancy in breast cancer, as well as to highlight the undergoing research in the development of angiogenesis-targeting therapies.
Collapse
Affiliation(s)
- Chikezie O. Madu
- Departments of Biological Sciences, University of Memphis, Memphis, TN 38152. USA
| | - Stephanie Wang
- Departments of Biology and Advanced Placement Biology, White Station High School, Memphis, TN 38117. USA
| | - Chinua O. Madu
- Departments of Biology and Advanced Placement Biology, White Station High School, Memphis, TN 38117. USA
| | - Yi Lu
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, Memphis, TN 38163. USA
| |
Collapse
|
41
|
Bloom MJ, Jarrett AM, Triplett TA, Syed AK, Davis T, Yankeelov TE, Sorace AG. Anti-HER2 induced myeloid cell alterations correspond with increasing vascular maturation in a murine model of HER2+ breast cancer. BMC Cancer 2020; 20:359. [PMID: 32345237 PMCID: PMC7189470 DOI: 10.1186/s12885-020-06868-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/14/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Therapy targeted to the human epidermal growth factor receptor type 2 (HER2) is used in combination with cytotoxic therapy in treatment of HER2+ breast cancer. Trastuzumab, a monoclonal antibody that targets HER2, has been shown pre-clinically to induce vascular changes that can increase delivery of chemotherapy. To quantify the role of immune modulation in treatment-induced vascular changes, this study identifies temporal changes in myeloid cell infiltration with corresponding vascular alterations in a preclinical model of HER2+ breast cancer following trastuzumab treatment. METHODS HER2+ tumor-bearing mice (N = 46) were treated with trastuzumab or saline. After extraction, half of each tumor was analyzed by immunophenotyping using flow cytometry. The other half was quantified by immunohistochemistry to characterize macrophage infiltration (F4/80), vascularity (CD31 and α-SMA), proliferation (Ki67) and cellularity (H&E). Additional mice (N = 10) were used to quantify differences in tumor cytokines between control and treated groups. RESULTS Immunophenotyping showed an increase in macrophage infiltration 24 h after trastuzumab treatment (P ≤ 0.05). With continued trastuzumab treatment, the M1 macrophage population increased (P = 0.02). Increases in vessel maturation index (i.e., the ratio of α-SMA to CD31) positively correlated with increases in tumor infiltrating M1 macrophages (R = 0.33, P = 0.04). Decreases in VEGF-A and increases in inflammatory cytokines (TNF-α, IL-1β, CCL21, CCL7, and CXCL10) were observed with continued trastuzumab treatment (P ≤ 0.05). CONCLUSIONS Preliminary results from this study in a murine model of HER2+ breast cancer show correlations between immune modulation and vascular changes, and reveals the potential for anti-HER2 therapy to reprogram immunosuppressive components of the tumor microenvironment. The quantification of immune modulation in HER2+ breast cancer, as well as the mechanistic insight of vascular alterations after anti-HER2 treatment, represent novel contributions and warrant further assessment for potential clinical translation.
Collapse
Affiliation(s)
- Meghan J Bloom
- Department of Biomedical Engineering, The University of Texas, Austin, TX, USA
| | - Angela M Jarrett
- LiveSTRONG Cancer Institutes, The University of Texas, Austin, TX, USA
| | - Todd A Triplett
- LiveSTRONG Cancer Institutes, The University of Texas, Austin, TX, USA.,Department of Oncology, The University of Texas Dell Medical School, Austin, TX, USA
| | - Anum K Syed
- Department of Biomedical Engineering, The University of Texas, Austin, TX, USA
| | - Tessa Davis
- Department of Biomedical Engineering, The University of Texas, Austin, TX, USA
| | - Thomas E Yankeelov
- Department of Biomedical Engineering, The University of Texas, Austin, TX, USA.,LiveSTRONG Cancer Institutes, The University of Texas, Austin, TX, USA.,Department of Oncology, The University of Texas Dell Medical School, Austin, TX, USA.,Diagnostic Medicine, The University of Texas, Austin, TX, USA.,Oden Institute for Computational and Engineering Sciences, The University of Texas, Austin, TX, USA
| | - Anna G Sorace
- Department of Radiology, The University of Alabama, Birmingham, AL, USA. .,Department of Biomedical Engineering, The University of Alabama, Birmingham, AL, USA. .,O'Neal Comprehensive Cancer Center, The University of Alabama, Birmingham, AL, USA.
| |
Collapse
|
42
|
Danni L, Lingyun Z, Jian W, Hongfei Y, Lu X, Peng Y, Xiujuan Q, Yunpeng L, Yuee T. Significant response to apatinib monotherapy in heavily pretreated advanced HER2-positive breast cancer: a case report and literature review. Cancer Biol Ther 2020; 21:590-596. [PMID: 32233990 DOI: 10.1080/15384047.2020.1743159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Although HER2-targeted therapy has been shown to prolong the survival of patients with HER2-positive breast cancer, most patients eventually progress due to drug resistance. Novel treatment options are urgently needed to overcome resistance to HER2-targeted therapy. The VEGF/VEGFR (Vascular endothelial growth factor and its receptors) pathway is essential in tumor angiogenesis, which may be a promising target in HER2-positive breast cancer providing a rationale for the use of tyrosine kinase inhibitors (TKIs) targeting VEGFR. Here, we present a case of a heavily pretreated advanced breast cancer patient who did not respond to HER2-targeted therapy and developed resistance to multiple lines of HER2-targeted treatment. The patient was treated with apatinib at a dose of 500 mg daily, and obtained partial remission (PR) with a progression-free-stage (PFS) of 6 months. Our case indicates that apatinib might have anti-tumor activity in patients with HER2-positive breast cancer with HER2-targeted resistance. This case is of value which may provide new insights into strategies for HER2-targeted therapy resistance options in the clinic.
Collapse
Affiliation(s)
- Li Danni
- Department of Medical Oncology, The First Hospital of China Medical University , Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University , Shenyang, China.,Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University , Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, the First Hospital of China Medical University , Shenyang, China
| | - Zhang Lingyun
- Department of Medical Oncology, The First Hospital of China Medical University , Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University , Shenyang, China.,Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University , Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, the First Hospital of China Medical University , Shenyang, China
| | - Wang Jian
- Department of Pathology, The First Hospital of China Medical University , Shenyang, China
| | - Yan Hongfei
- Department of Medical Oncology, The First Hospital of China Medical University , Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University , Shenyang, China.,Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University , Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, the First Hospital of China Medical University , Shenyang, China
| | - Xu Lu
- Department of Medical Oncology, The First Hospital of China Medical University , Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University , Shenyang, China.,Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University , Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, the First Hospital of China Medical University , Shenyang, China
| | - Yang Peng
- Nanjing Geneseeq Technology Inc , Nanjing, China
| | - Qu Xiujuan
- Department of Medical Oncology, The First Hospital of China Medical University , Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University , Shenyang, China.,Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University , Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, the First Hospital of China Medical University , Shenyang, China
| | - Liu Yunpeng
- Department of Medical Oncology, The First Hospital of China Medical University , Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University , Shenyang, China.,Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University , Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, the First Hospital of China Medical University , Shenyang, China
| | - Teng Yuee
- Department of Medical Oncology, The First Hospital of China Medical University , Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University , Shenyang, China.,Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University , Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, the First Hospital of China Medical University , Shenyang, China
| |
Collapse
|
43
|
Hey SP, Gyawali B, D’Andrea E, Kanagaraj M, Franklin JM, Kesselheim AS. A Systematic Review and Meta-Analysis of Bevacizumab in First-Line Metastatic Breast Cancer: Lessons for Research and Regulatory Enterprises. J Natl Cancer Inst 2020; 112:335-342. [PMID: 31651981 PMCID: PMC7156929 DOI: 10.1093/jnci/djz211] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 10/14/2019] [Accepted: 10/23/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND The US Food and Drug Administration's accelerated approval and later withdrawal of bevacizumab in patients with metastatic breast cancer (mBC) is a seminal case for ongoing debates about the validity of using progression-free survival (PFS) as a surrogate measure for overall survival (OS) in cancer drug approvals. We systematically reviewed and meta-analyzed the evidence around bevacizumab's regulatory approval and withdrawal in mBC. METHODS We searched for all published phase II or III clinical trials testing bevacizumab as a first-line therapy for patients with mBC. Data were extracted on trial demographics, interventions, and outcomes. Descriptive analysis was stratified by whether the trial was initiated before, during, or after the accelerated approval. We used a cumulative random-effects meta-analysis to assess the evolution of evidence of the effect of bevacizumab on PFS and OS. We estimated the association between the trial-level PFS and OS effect using a nonlinear mixed-regression model. RESULTS Fifty-two studies were included. Trial activity dramatically dropped after the accelerated approval was withdrawn. Eight clinical trials reported hazard ratios (hazard ratios) and were meta-analyzed. The cumulative hazard ratio for PFS was 0.72 (95% CI = 0.65 to 0.79), and the cumulative hazard ratio for OS was 0.90 (95% CI = 0.80 to 1.01). The regression model showed a statistically nonsignificant association between PFS benefit and OS benefit (β = 0.43, SE = 0.81). CONCLUSION The US Food and Drug Administration's decision-making in this case was consistent with the evolving state of evidence. However, the fact that seven clinical trials are insufficient to conclude validity (or lack thereof) for a trial-level surrogate suggests that it would be more efficient to conduct trials using the more clinically meaningful endpoints.
Collapse
Affiliation(s)
- Spencer Phillips Hey
- Program on Regulation, Therapeutics, and Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
- Harvard Center for Bioethics, Harvard Medical School, Boston, MA
| | - Bishal Gyawali
- Program on Regulation, Therapeutics, and Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
- Department of Oncology, Department of Public Health Sciences, and Division of Cancer Care and Epidemiology, Queen’s University, Kingston, Ontario, Canada
| | - Elvira D’Andrea
- Program on Regulation, Therapeutics, and Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | | | - Jessica M Franklin
- Program on Regulation, Therapeutics, and Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Aaron S Kesselheim
- Program on Regulation, Therapeutics, and Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
- Harvard Center for Bioethics, Harvard Medical School, Boston, MA
| |
Collapse
|
44
|
Costa RLB, Czerniecki BJ. Clinical development of immunotherapies for HER2 + breast cancer: a review of HER2-directed monoclonal antibodies and beyond. NPJ Breast Cancer 2020; 6:10. [PMID: 32195333 PMCID: PMC7067811 DOI: 10.1038/s41523-020-0153-3] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 02/18/2020] [Indexed: 02/07/2023] Open
Abstract
Human epidermal growth factor receptor 2-positive (HER2+) breast cancer accounts for ~25% of breast cancer cases. Monoclonal antibodies (mAbs) against HER2 have led to unparalleled clinical benefit for a subset of patients with HER2+ breast cancer. In this narrative review, we summarize advances in the understanding of immune system interactions, examine clinical developments, and suggest rationales for future investigation of immunotherapies for HER2+ breast cancer. Complex interactions have been found between different branches of the immune system, HER2+ breast cancer, and targeted treatments (approved and under investigation). A new wave of immunotherapies, such as novel HER2-directed mAbs, antibody drug conjugates, vaccines, and adoptive T-cell therapies, are being studied in a broad population of patients with HER2-expressing tumors. The development of immunotherapies for HER2+ breast cancer represents an evolving field that should take into account interactions between different components of the immune system.
Collapse
Affiliation(s)
- Ricardo L B Costa
- Departments of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL USA
| | - Brian J Czerniecki
- Departments of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL USA
| |
Collapse
|
45
|
Larbouret C, Poul MA, Chardès T. [Mimicking polyclonal immune response in therapy: from combination of two monoclonal antibodies to oligoclonal antibody-based mixtures]. Med Sci (Paris) 2020; 35:1083-1091. [PMID: 31903921 DOI: 10.1051/medsci/2019216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Monoclonal antibodies have revolutionized the treatment of many diseases, but their clinical effectiveness remains limited in some cases. Associations of antibodies binding to the same target (homo-combination) or to several different targets (hetero-combination), thereby mimicking a polyclonal humoral immune response, have demonstrated a therapeutic improvement in pre-clinical and clinical trials, mainly in the field of oncology and infectious diseases. The combinations increase the efficacy of the biological responses and override resistance mechanisms observed with antibody monotherapy. The most common method of formulating and administering antibody combinations is a separate formulation, with sequential injection of each antibody as individual drug substance. Alternatively, combined formulations are developed where the separately-produced antibodies are mixed before administration or produced simultaneously by a single cell line, or a mixture of cell lines as a polyclonal master cell bank. The regulation, the toxicity and the injection sequence of these oligoclonal antibody-based mixtures remain points to be clarified and optimized for a better therapeutic effect.
Collapse
Affiliation(s)
- Christel Larbouret
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université de Montpellier, Institut régional du Cancer de Montpellier (ICM), 34298 Montpellier, France
| | - Marie-Alix Poul
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université de Montpellier, Institut régional du Cancer de Montpellier (ICM), 34298 Montpellier, France
| | - Thierry Chardès
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université de Montpellier, Institut régional du Cancer de Montpellier (ICM), 34298 Montpellier, France - Centre National de la Recherche Scientifique (CNRS), Paris, France
| |
Collapse
|
46
|
Lamb LS, Sim HW, McCormack AI. Case Report: A Case of Pituitary Carcinoma Treated With Sequential Dual Immunotherapy and Vascular Endothelial Growth Factor Inhibition Therapy. Front Endocrinol (Lausanne) 2020; 11:576027. [PMID: 33312158 PMCID: PMC7708326 DOI: 10.3389/fendo.2020.576027] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 10/19/2020] [Indexed: 12/18/2022] Open
Abstract
Aggressive pituitary tumors (APTs) are associated with significant morbidity and mortality, and effective treatment options are limited. Immune checkpoint inhibitors (ICIs) have revolutionized clinical cancer care; however, there is little experience with these agents in the management of APTs. Vascular endothelial growth factor (VEGF) targeted therapy has reported success in a small number of APT case reports. Here we describe a case of pituitary carcinoma responding to ICI therapy and subsequently VEGF inhibition. We discuss the possible mechanisms and experience with ICI therapy and VEGF inhibitors in the management of APTs, biomarkers that may predict response, and the potential role of combination therapies including ICIs and temozolomide.
Collapse
Affiliation(s)
- Lydia S. Lamb
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- St Vincent’s Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Hao-Wen Sim
- St Vincent’s Clinical School, University of New South Wales, Sydney, NSW, Australia
- The Kinghorn Cancer Centre, Sydney, NSW, Australia
| | - Ann I. McCormack
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- St Vincent’s Clinical School, University of New South Wales, Sydney, NSW, Australia
- Department of Endocrinology, St Vincent’s Hospital, Sydney, NSW, Australia
- *Correspondence: Ann I. McCormack,
| |
Collapse
|
47
|
Vessel co-option and resistance to anti-angiogenic therapy. Angiogenesis 2019; 23:55-74. [PMID: 31865479 DOI: 10.1007/s10456-019-09698-6] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/22/2019] [Indexed: 12/20/2022]
Abstract
Vessel co-option is a non-angiogenic mechanism of tumour vascularisation in which cancer cells utilise pre-existing blood vessels instead of inducing new blood vessel formation. Vessel co-option has been observed across a range of different tumour types, in both primary cancers and metastatic disease. Importantly, vessel co-option is now implicated as a major mechanism that mediates resistance to conventional anti-angiogenic drugs and this may help to explain the limited efficacy of this therapeutic approach in certain clinical settings. This includes the use of anti-angiogenic drugs to treat advanced-stage/metastatic disease, treatment in the adjuvant setting and the treatment of primary disease. In this article, we review the available evidence linking vessel co-option with resistance to anti-angiogenic therapy in numerous tumour types, including breast, colorectal, lung and pancreatic cancer, glioblastoma, melanoma, hepatocellular carcinoma, and renal cell carcinoma. The finding that vessel co-option is a significant mechanism of resistance to anti-angiogenic therapy may have important implications for the future of anti-cancer therapy, including (a) predicting response to anti-angiogenic drugs, (b) the need to develop therapies that target both angiogenesis and vessel co-option in tumours, and (c) predicting the response to other therapeutic modalities, including immunotherapy.
Collapse
|
48
|
Guarischi-Sousa R, Monteiro JS, Alecrim LC, Michaloski JS, Cardeal LB, Ferreira EN, Carraro DM, Nunes DN, Dias-Neto E, Reimand J, Boutros PC, Setubal JC, Giordano RJ. A transcriptome-based signature of pathological angiogenesis predicts breast cancer patient survival. PLoS Genet 2019; 15:e1008482. [PMID: 31846472 PMCID: PMC6917213 DOI: 10.1371/journal.pgen.1008482] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 10/15/2019] [Indexed: 12/30/2022] Open
Abstract
The specific genes and molecules that drive physiological angiogenesis differ from those involved in pathological angiogenesis, suggesting distinct mechanisms for these seemingly related processes. Unveiling genes and pathways preferentially associated with pathologic angiogenesis is key to understanding its mechanisms, thereby facilitating development of novel approaches to managing angiogenesis-dependent diseases. To better understand these different processes, we elucidated the transcriptome of the mouse retina in the well-accepted oxygen-induced retinopathy (OIR) model of pathological angiogenesis. We identified 153 genes changed between normal and OIR retinas, which represent a molecular signature relevant to other angiogenesis-dependent processes such as cancer. These genes robustly predict the survival of breast cancer patients, which was validated in an independent 1,000-patient test cohort (40% difference in 15-year survival; p = 2.56 x 10-21). These results suggest that the OIR model reveals key genes involved in pathological angiogenesis, and these may find important applications in stratifying tumors for treatment intensification or for angiogenesis-targeted therapies.
Collapse
Affiliation(s)
- Rodrigo Guarischi-Sousa
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Jhonatas S. Monteiro
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Lilian C. Alecrim
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Jussara S. Michaloski
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Laura B. Cardeal
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Elisa N. Ferreira
- International Research Center (CIPE) A.C. Camargo Cancer Center, São Paulo, SP, Brazil
| | - Dirce M. Carraro
- International Research Center (CIPE) A.C. Camargo Cancer Center, São Paulo, SP, Brazil
| | - Diana N. Nunes
- International Research Center (CIPE) A.C. Camargo Cancer Center, São Paulo, SP, Brazil
| | - Emmanuel Dias-Neto
- International Research Center (CIPE) A.C. Camargo Cancer Center, São Paulo, SP, Brazil
- Laboratory of Neurosciences (LIM27), Institute & Department of Psychiatry, University of São Paulo, São Paulo, Brazil
| | - Jüri Reimand
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Paul C. Boutros
- Department of Human Genetics, University of California Los Angeles (UCLA), Los Angeles, CA, United States of America
- * E-mail: (PCB); (JCS); (RJG)
| | - João C. Setubal
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
- * E-mail: (PCB); (JCS); (RJG)
| | - Ricardo J. Giordano
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
- * E-mail: (PCB); (JCS); (RJG)
| |
Collapse
|
49
|
Kuczynski EA, Vermeulen PB, Pezzella F, Kerbel RS, Reynolds AR. Vessel co-option in cancer. Nat Rev Clin Oncol 2019; 16:469-493. [PMID: 30816337 DOI: 10.1038/s41571-019-0181-9] [Citation(s) in RCA: 281] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
All solid tumours require a vascular supply in order to progress. Although the ability to induce angiogenesis (new blood vessel growth) has long been regarded as essential to this purpose, thus far, anti-angiogenic therapies have shown only modest efficacy in patients. Importantly, overshadowed by the literature on tumour angiogenesis is a long-standing, but continually emerging, body of research indicating that tumours can grow instead by hijacking pre-existing blood vessels of the surrounding nonmalignant tissue. This process, termed vessel co-option, is a frequently overlooked mechanism of tumour vascularization that can influence disease progression, metastasis and response to treatment. In this Review, we describe the evidence that tumours located at numerous anatomical sites can exploit vessel co-option. We also discuss the proposed molecular mechanisms involved and the multifaceted implications of vessel co-option for patient outcomes.
Collapse
Affiliation(s)
- Elizabeth A Kuczynski
- Bioscience, Oncology, IMED Biotech Unit, AstraZeneca, Cambridge, UK. .,Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Canada.
| | - Peter B Vermeulen
- HistoGeneX, Antwerp, Belgium.,Translational Cancer Research Unit, GZA Hospitals St Augustinus, University of Antwerp, Wilrijk-Antwerp, Belgium.,Tumour Biology Team, Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Francesco Pezzella
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Robert S Kerbel
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Andrew R Reynolds
- Tumour Biology Team, Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK. .,Oncology Translational Medicine Unit, IMED Biotech Unit, AstraZeneca, Cambridge, UK.
| |
Collapse
|
50
|
Xie BJ, Zhu LN, Ma C, Li JB, Dong L, Zhu ZN, Ding T, Gu XS. A network meta-analysis on the efficacy of HER2-targeted agents in combination with taxane-containing regimens for treatment of HER2-positive metastatic breast cancer. Breast Cancer 2019; 27:186-196. [DOI: 10.1007/s12282-019-01007-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 08/27/2019] [Indexed: 12/28/2022]
|