151
|
Rizzo A, Schipilliti FM, Di Costanzo F, Acquafredda S, Arpino G, Puglisi F, Del Mastro L, Montemurro F, De Laurentiis M, Giuliano M. Discontinuation rate and serious adverse events of chemoimmunotherapy as neoadjuvant treatment for triple-negative breast cancer: a systematic review and meta-analysis. ESMO Open 2023; 8:102198. [PMID: 38100933 PMCID: PMC10774952 DOI: 10.1016/j.esmoop.2023.102198] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND The use of combination of chemotherapy with immune checkpoint inhibitors (ICIs) has shown efficacy in triple-negative breast cancer (TNBC), and chemoimmunotherapy has been introduced in clinical practice. However, limited data are available on the discontinuation rate and serious adverse events of these treatments, particularly in the neoadjuvant setting. Herein, we carried out a comprehensive systematic review and meta-analysis to assess discontinuation rate and serious adverse events of chemoimmunotherapy compared to chemotherapy alone in phase II and III neoadjuvant clinical trials in TNBC. MATERIALS AND METHODS Following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, EMBASE, Cochrane Library, and PubMed/Medline were searched for articles published from June 2008 to May 2023. The outcomes of interest were the discontinuation rate, serious adverse events, and grade 3-4 adverse events. RESULTS Four studies were included in the analysis. The pooled odds ratios (ORs) for discontinuation rate and serious adverse events were 1.26 [95% confidence interval (CI) 0.78-2.06] and 1.79 (95% CI 1.4-2.28), respectively, in patients receiving chemoimmunotherapy compared to chemotherapy alone as neoadjuvant treatment for TNBC. The chemoimmunotherapy group had a higher risk of grade 3-4 adverse events (OR 1.30, 95% CI 1.07-1.59). The analysis showed substantial heterogeneity, and the risk of discontinuation rate was heavily influenced by the KEYNOTE-522 trial. CONCLUSIONS Our findings highlight the need for clinical trials specifically focused on safety, quality of life, and treatment adherence in TNBC patients receiving neoadjuvant treatment. Close monitoring of tolerability remains crucial in this clinical setting.
Collapse
Affiliation(s)
- A Rizzo
- IRCCS Istituto Tumori "Giovanni Paolo II", Bari
| | - F M Schipilliti
- Oncological Department, Sant'Andrea Hospital, University Sapienza in Rome, Rome
| | - F Di Costanzo
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples
| | | | - G Arpino
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples
| | - F Puglisi
- Department of Medicine, University of Udine, Udine; Department of Medical Oncology-CRO Aviano, National Cancer Institute, IRCCS, Aviano
| | - L Del Mastro
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova; Medical Oncology Department, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova
| | | | - M De Laurentiis
- Istituto Nazionale Tumori IRCCS "Fondazione Pascale", Napoli, Italy
| | - M Giuliano
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples.
| |
Collapse
|
152
|
Wong RSJ, Ong RJM, Lim JSJ. Immune checkpoint inhibitors in breast cancer: development, mechanisms of resistance and potential management strategies. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:768-787. [PMID: 38263984 PMCID: PMC10804393 DOI: 10.20517/cdr.2023.58] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 10/14/2023] [Accepted: 10/31/2023] [Indexed: 01/25/2024]
Abstract
The use of immune checkpoint inhibitors (ICIs) has increased exponentially in the past decade, although its progress specifically for breast cancer has been modest. The first U.S. Food and Drug Administration approval for ICI in breast cancer came in 2019, eight years after the first-ever approval of an ICI. At present, current indications for ICIs are relevant only to a subset of patients with triple-negative breast cancer, or those displaying high microsatellite instability or deficiency in the mismatch repair protein pathway. With an increasing understanding of the limitations of using ICIs, which stem from breast cancer being innately poorly immunogenic, as well as the presence of various intrinsic and acquired resistance pathways, ongoing trials are evaluating different combination therapies to overcome these barriers. In this review, we aim to describe the development timeline of ICIs and resistance mechanisms limiting their utility, and summarise the available approaches and ongoing trials relevant to overcoming each resistance mechanism.
Collapse
Affiliation(s)
- Rachel SJ Wong
- Department of Haematology-Oncology, National University Cancer Institute, National University Hospital, Singapore 119228, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Rebecca JM Ong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Joline SJ Lim
- Department of Haematology-Oncology, National University Cancer Institute, National University Hospital, Singapore 119228, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| |
Collapse
|
153
|
Pescia C, Guerini-Rocco E, Viale G, Fusco N. Advances in Early Breast Cancer Risk Profiling: From Histopathology to Molecular Technologies. Cancers (Basel) 2023; 15:5430. [PMID: 38001690 PMCID: PMC10670146 DOI: 10.3390/cancers15225430] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/05/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Early breast cancer (BC) is the definition applied to breast-confined tumors with or without limited involvement of locoregional lymph nodes. While risk stratification is essential for guiding clinical decisions, it can be a complex endeavor in these patients due to the absence of comprehensive guidelines. Histopathological analysis and biomarker assessment play a pivotal role in defining patient outcomes. Traditional histological criteria such as tumor size, lymph node involvement, histological type and grade, lymphovascular invasion, and immune cell infiltration are significant prognostic indicators. In addition to the hormone receptor, HER2, and-in specific scenarios-BRCA1/2 testing, molecular subtyping through gene expression profiling provides valuable insights to tailor clinical decision-making. The emergence of "omics" technologies, applicable to both tissue and liquid biopsy samples, has broadened our arsenal for evaluating the risk of early BC. However, a pressing need remains for standardized methodologies and integrated pathological models that encompass multiple analytical dimensions. In this study, we provide a detailed examination of the existing strategies for early BC risk stratification, intending to serve as a practical guide for histopathologists and molecular pathologists.
Collapse
Affiliation(s)
- Carlo Pescia
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (C.P.); (E.G.-R.); (G.V.)
- School of Pathology, University of Milan, 20141 Milan, Italy
| | - Elena Guerini-Rocco
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (C.P.); (E.G.-R.); (G.V.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy
| | - Giuseppe Viale
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (C.P.); (E.G.-R.); (G.V.)
| | - Nicola Fusco
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (C.P.); (E.G.-R.); (G.V.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy
| |
Collapse
|
154
|
Wimmer K, Sachet M, Ramos C, Frantal S, Birnleitner H, Brostjan C, Exner R, Filipits M, Bago-Horvath Z, Rudas M, Bartsch R, Gnant M, Singer CF, Balic M, Egle D, Oehler R, Fitzal F. Differential immunomodulatory effects of epirubicin/cyclophosphamide and docetaxel in breast cancer patients. J Exp Clin Cancer Res 2023; 42:300. [PMID: 37957750 PMCID: PMC10644559 DOI: 10.1186/s13046-023-02876-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] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 10/29/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND Epirubicin/cyclophosphamide (EC) and docetaxel (D) are commonly used in a sequential regimen in the neoadjuvant treatment of early, high-risk or locally advanced breast cancer (BC). Novel approaches to increase the response rate combine this treatment with immunotherapies such as PD-1 inhibition. However, the expected stimulatory effect on lymphocytes may depend on the chemotherapy backbone. Therefore, we separately compared the immunomodulatory effects of EC and D in the setting of a randomized clinical trial. METHODS Tumor and blood samples of 154 patients from the ABCSG-34 trial were available (76 patients received four cycles of EC followed by four cycles of D; 78 patients get the reverse treatment sequence). Tumor-infiltrating lymphocytes, circulating lymphocytes and 14 soluble immune mediators were determined at baseline and at drug change. Furthermore, six BC cell lines were treated with E, C or D and co-cultured with immune cells. RESULTS Initial treatment with four cycles of EC reduced circulating B and T cells by 94% and 45%, respectively. In contrast, no comparable effects on lymphocytes were observed in patients treated with initial four cycles of D. Most immune mediators decreased under EC whereas D-treatment resulted in elevated levels of CXCL10, urokinase-type plasminogen activator (uPA) and its soluble receptor (suPAR). Accordingly, only the exposure of BC cell lines to D induced similar increases as compared to E. While treatment of BC cells with E was associated with cell shrinkage and apoptosis, D induced cell swelling and accumulation of cells in G2 phase. CONCLUSION The deleterious effect of EC on lymphocytes indicates strong immunosuppressive properties of this combination therapy. D, in contrast, has no effect on lymphocytes, but triggers the secretion of stimulatory proteins in vivo and in vitro, indicating a supportive effect on the immune system. Underlying differences in the induced cell death might be causal. These divergent immunomodulatory effects of epirubicin/cyclophosphamide and docetaxel should be considered when planning future combinations with immunotherapies in breast cancer.
Collapse
Affiliation(s)
- Kerstin Wimmer
- Department of General Surgery, Division of Visceral Surgery and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
| | - Monika Sachet
- Department of General Surgery, Division of Visceral Surgery and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| | - Cristiano Ramos
- Department of General Surgery, Division of Visceral Surgery and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| | - Sophie Frantal
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
| | - Hanna Birnleitner
- Department of General Surgery, Division of Visceral Surgery and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| | - Christine Brostjan
- Department of General Surgery, Division of Vascular Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Ruth Exner
- Department of General Surgery, Division of Visceral Surgery and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| | - Martin Filipits
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Center for Cancer Research, Medical University of Vienna, 1090, Vienna, Austria
| | - Zsuzsanna Bago-Horvath
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Department of Pathology, Medical University of Vienna, 1090, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Margaretha Rudas
- Department of Pathology, Medical University of Vienna, 1090, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Rupert Bartsch
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Department of Medicine 1, Division of Oncology, Medical University of Vienna, 1090, Vienna, Austria
| | - Michael Gnant
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Christian F Singer
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
- Department of Gynecology, Medical University of Vienna, 1090, Vienna, Austria
| | - Marija Balic
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Department of Oncology, Medical University of Graz, Graz, Austria
| | - Daniel Egle
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Department of Gynecology, Medical University Innsbruck, Innsbruck, Austria
| | - Rudolf Oehler
- Department of General Surgery, Division of Visceral Surgery and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria.
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria.
| | - Florian Fitzal
- Department of General Surgery, Division of Visceral Surgery and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
| |
Collapse
|
155
|
Roy AM, Patel A, Catalfamo K, Attwood K, Khoury T, Yao S, Gandhi S. Racial and Ethnic Disparity in Preoperative Chemosensitivity and Survival in Patients With Early-Stage Breast Cancer. JAMA Netw Open 2023; 6:e2344517. [PMID: 37991763 PMCID: PMC10665980 DOI: 10.1001/jamanetworkopen.2023.44517] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 10/05/2023] [Indexed: 11/23/2023] Open
Abstract
Importance It remains unclear what survival benefit is associated with preoperative chemosensitivity after receiving neoadjuvant chemotherapy (NACT) among patients with resectable breast cancer from diverse racial and ethnic backgrounds. Objective To investigate racial and ethnic disparities in chemosensitivity and association with survival in patients with early-stage breast cancer. Design, Setting, and Participants This retrospective cohort study queried data from the National Cancer Database (NCDB) between calendar years 2010 and 2018. Participants included patients with breast cancer with clinical stage I to III disease treated with NACT. Preoperative chemosensitivity was defined as very sensitive (ypT0N0), sensitive (pathologic TNM stage less than clinical stage, excluding ypT0N0), and refractory (pathologic stage greater than or equal to clinical stage). Data were analyzed in November 2022. Exposure Receipt of NACT and clinicopathologic and treatment factors contributing to racial and ethnic disparities in survival. Main Outcomes and Measures Overall survival of patients from diverse racial and ethnic backgrounds who received NACT. Results This study included 103 605 patients (median age, 53 [IQR, 44-62] years, 99.5% [n = 103 060] women, and 68.7% [n = 71 203] White race). Among them, breast cancer was refractory in 43.2% (n = 44 796), sensitive in 34.4% (n = 35 638), and very sensitive in 22.4% (n = 23 171) of patients. In the hormone receptor-positive ERBB2 negative (formerly HER2 negative) group, patients had more refractory disease regardless of race or ethnicity (all races and ethnicities refractory: 54%-59%; P < .001). Among ERBB2 positive disease, Black patients had a lower percentage of very sensitive disease (32% vs 37%-40%; P < .001) and among triple-negative breast cancer, more refractory disease was seen among Black patients compared with other races and ethnicities (38% vs 30%-35%; P < .001). In refractory (hazard ratio [HR], 1.53; 95% CI, 1.47-1.60; P < .001) and sensitive (HR, 1.25; 95% CI, 1.17-1.33; P < .001) disease, Black patients had a higher mortality risk compared with White patients in the overall cohort. Asian patients had a lower mortality risk compared with White patients in refractory (HR, 0.71; 95% CI, 0.63-0.80; P < .001), sensitive (HR, 0.58; 95% CI, 0.49-0.69; P < .001), and very sensitive (HR, 0.60; 95% CI, 0.43-0.82; P < .001) disease groups in the overall cohort. Conclusions and Relevance In this cohort study, Black patients had a higher mortality risk compared with White patients among those with residual disease after NACT. This highlights the need for personalized treatment strategies for Black patients to help them attain pathologic complete response.
Collapse
Affiliation(s)
- Arya Mariam Roy
- Department of Hematology and Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Archit Patel
- Department of Hematology and Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Kayla Catalfamo
- Department of Biostatistics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Kristopher Attwood
- Department of Biostatistics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Thaer Khoury
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Song Yao
- Department of Cancer Prevention & Control, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Shipra Gandhi
- Department of Hematology and Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| |
Collapse
|
156
|
Seban RD, Arnaud E, Loirat D, Cabel L, Cottu P, Djerroudi L, Hescot S, Loap P, Bonneau C, Bidard FC, Huchet V, Jehanno N, Berenbaum A, Champion L, Buvat I. [18F]FDG PET/CT for predicting triple-negative breast cancer outcomes after neoadjuvant chemotherapy with or without pembrolizumab. Eur J Nucl Med Mol Imaging 2023; 50:4024-4035. [PMID: 37606858 DOI: 10.1007/s00259-023-06394-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/08/2023] [Indexed: 08/23/2023]
Abstract
PURPOSE To determine if pretreatment [18F]FDG PET/CT could contribute to predicting complete pathological complete response (pCR) in patients with early-stage triple-negative breast cancer (TNBC) undergoing neoadjuvant chemotherapy with or without pembrolizumab. METHODS In this retrospective bicentric study, we included TNBC patients who underwent [18F]FDG PET/CT before neoadjuvant chemotherapy (NAC) or chemo-immunotherapy (NACI) between March 2017 and August 2022. Clinical, biological, and pathological data were collected. Tumor SUVmax and total metabolic tumor volume (TMTV) were measured from the PET images. Cut-off values were determined using ROC curves and a multivariable model was developed using logistic regression to predict pCR. RESULTS N = 191 patients were included. pCR rates were 53 and 70% in patients treated with NAC (N = 91) and NACI (N = 100), respectively (p < 0.01). In univariable analysis, high Ki67, high tumor SUVmax (> 12.3), and low TMTV (≤ 3.0 cm3) were predictors of pCR in the NAC cohort while tumor staging classification (< T3), BRCA1/2 germline mutation, high tumor SUVmax (> 17.2), and low TMTV (≤ 7.3 cm3) correlated with pCR in the NACI cohort. In multivariable analysis, only high tumor SUVmax (NAC: OR 8.8, p < 0.01; NACI: OR 3.7, p = 0.02) and low TMTV (NAC: OR 6.6, p < 0.01; NACI: OR 3.5, p = 0.03) were independent factors for pCR in both cohorts, albeit at different thresholds. CONCLUSION High tumor metabolism (SUVmax) and low tumor burden (TMTV) could predict pCR after NAC regardless of the addition of pembrolizumab. Further studies are warranted to validate such findings and determine how these biomarkers could be used to guide neoadjuvant therapy in TNBC patients.
Collapse
Affiliation(s)
- Romain-David Seban
- Department of Nuclear Medicine and Endocrine Oncology, Institut Curie, 92210, Saint-Cloud, France.
- Laboratoire d'Imagerie Translationnelle en Oncologie, Inserm U1288, Institut Curie, PSL University, Paris Saclay University, 91400, Orsay, France.
| | - Emilie Arnaud
- Department of Medical Oncology, Institut Curie, PSL Research University, 75005, Paris, France
| | - Delphine Loirat
- Department of Medical Oncology, Institut Curie, PSL Research University, 75005, Paris, France
| | - Luc Cabel
- Department of Medical Oncology, Institut Curie, PSL Research University, 75005, Paris, France
| | - Paul Cottu
- Department of Medical Oncology, Institut Curie, PSL Research University, 75005, Paris, France
| | | | - Segolene Hescot
- Department of Nuclear Medicine and Endocrine Oncology, Institut Curie, 92210, Saint-Cloud, France
| | - Pierre Loap
- Department of Radiation Oncology, Institut Curie, 92210, Saint-Cloud, France
| | - Claire Bonneau
- Inserm U900, Institut Curie, 35, rue Dailly, 92210, Saint-Cloud, France
- Department of Surgery, Institut Curie, 92210, Saint-Cloud, France
| | - Francois-Clement Bidard
- Department of Medical Oncology, Institut Curie, UVSQ/Paris-Saclay University, 92210, Saint-Cloud, France
- Circulating Tumor Biomarkers Laboratory, Institut Curie, SiRIC, PSL Research University, Paris, France
| | - Virginie Huchet
- Department of Nuclear Medicine, Institut Curie, 75005, Paris, France
| | - Nina Jehanno
- Department of Nuclear Medicine, Institut Curie, 75005, Paris, France
| | - Arnaud Berenbaum
- Department of Nuclear Medicine and Endocrine Oncology, Institut Curie, 92210, Saint-Cloud, France
| | - Laurence Champion
- Department of Nuclear Medicine and Endocrine Oncology, Institut Curie, 92210, Saint-Cloud, France
- Laboratoire d'Imagerie Translationnelle en Oncologie, Inserm U1288, Institut Curie, PSL University, Paris Saclay University, 91400, Orsay, France
| | - Irene Buvat
- Laboratoire d'Imagerie Translationnelle en Oncologie, Inserm U1288, Institut Curie, PSL University, Paris Saclay University, 91400, Orsay, France
| |
Collapse
|
157
|
Quan R, Li Z, Zhang H, Li X. Do patients receive any benefit from the addition of perioperative immunotherapy-chemotherapy for solid tumors? Medicine (Baltimore) 2023; 102:e35603. [PMID: 37861545 PMCID: PMC10589575 DOI: 10.1097/md.0000000000035603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 09/20/2023] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND Progress in the use of neoadjuvant immunotherapy combined with chemotherapy has become a highlight of cancer research. Our meta-analysis aimed to better elucidate the activity, efficacy and safety of this combination using data obtained from randomized controlled trials (RCTs). METHODS A systematic search of PubMed, Embase, Web of Science, the Cochrane Library and conference proceedings up to January 31, 2023 was carried out to identify RCTs investigating neoadjuvant immunotherapy combined with chemotherapy for the treatment of solid tumors. Using fixed- and random-effects models, pooled odds ratios (ORs) and hazard ratios with 95% confidence intervals (CIs) were calculated for pathological complete response (pCR, defined as ypT0/is pN0) and immunotherapy treatment-related adverse events. RESULTS A total of 1876 studies were identified, and 6 RCTs (N = 2558 patients) were included. The pCR was significantly higher with neoadjuvant immunotherapy combined with chemotherapy than with neoadjuvant chemotherapy alone (OR = 2.30, 95% CI: 1.43-3.71, P < .001). The pCR was confirmed to be statistically significant in the PD-L1-positive subgroup (OR = 2.01; 95% CI: 1.55-2.59, P = .012). The pCR was confirmed to be statistically significant in the PD-1 inhibitor subgroup (OR = 4.17; 95% CI: 1.47-11.87, P = .000), while no statistically significant was observed in the PD-L1 inhibitor subgroup (OR = 1.52; 95% CI: 1.12-2.07, P = .308). The pooled ORs of any grade treatment-related or immunotherapy-related adverse events were significant, but the grade 3-4 immunotherapy-related adverse events were not. CONCLUSION Our study provides comprehensive data that the addition of PD1 blockade to neoadjuvant chemotherapy resulted in better treatment efficacy than neoadjuvant chemotherapy alone in patients with solid tumors and had a similar safety profile.
Collapse
Affiliation(s)
- Rencui Quan
- Department of Radiation Oncology, Shenzhen People’s Hospital, The Second Clinic Medical College of Jinan University, Shenzhen, China
- First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Zaishang Li
- Department of Urology, Shenzhen People’s Hospital, The Second Clinic Medical College of Jinan University, Shenzhen, China
- Department of Urology, First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
- Department of Urology, Minimally Invasive Urology of Shenzhen Research and Development Center of Medical Engineering and Technology, Shenzhen, China
| | - Huaqing Zhang
- Department of Endocrinology, Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Xueying Li
- Department of Oncology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
158
|
Wang J, Wu SG. Breast Cancer: An Overview of Current Therapeutic Strategies, Challenge, and Perspectives. BREAST CANCER (DOVE MEDICAL PRESS) 2023; 15:721-730. [PMID: 37881514 PMCID: PMC10596062 DOI: 10.2147/bctt.s432526] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/11/2023] [Indexed: 10/27/2023]
Abstract
Breast cancer is the most commonly diagnosed cancer and the leading cause of death among female patients, which seriously threatens the health of women in the whole world. The treatments of breast cancer require the cooperation of a multidisciplinary setting and taking tumor load and molecular makers into account. For early breast cancer, breast-conserving surgery with radiotherapy or mastectomy alone remains the standard management, and the administration of adjuvant systemic therapy is decided by the status of lymph nodes, hormone receptors, and human epidermal growth factor receptor-2. For metastatic breast cancer, the goal of treatments is to prolong survival and maintain quality of life. This review will present the current advances and controversies of surgery, chemotherapy, radiotherapy, endocrine therapy, targeted therapy, immunotherapy, gene therapy, and other innovative treatment strategies in early-stage and metastatic breast cancer.
Collapse
Affiliation(s)
- Jun Wang
- Department of Head and Neck Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - San-Gang Wu
- Department of Radiation Oncology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, People’s Republic of China
| |
Collapse
|
159
|
Bhardwaj PV, Abdou YG. The Evolving Landscape of Immune Checkpoint Inhibitors and Antibody Drug Conjugates in the Treatment of Early-Stage Breast Cancer. Oncologist 2023; 28:832-844. [PMID: 37597245 PMCID: PMC11025387 DOI: 10.1093/oncolo/oyad233] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 07/24/2023] [Indexed: 08/21/2023] Open
Abstract
For decades, chemotherapy has been the mainstay of breast cancer treatment. Novel therapies are expanding the therapeutic options and altering the treatment algorithms to manage this disease. The use and approval of immune checkpoint inhibitors (ICIs) and antibody-drug conjugates (ADCs) represent a few areas of progress. These therapies initially gained attention in the metastatic setting but have subsequently found a role in early-stage breast cancer. Although human epidermal growth factor receptor 2 (HER2) is at the center of ADC development, other surface antigens with a differential expression between tumor and normal cells may be appropriate for ADC targeting. This has led to the discovery of new ADCs targeting other receptors, including TROP-2, HER-3, and LIV-1, to name a few. Similarly, the addition of pembrolizumab in treating early-stage triple-negative breast cancer has led to exploring other ICIs in this setting. However, it has also raised important scientific questions regarding optimal patient selection, biomarkers that predict the success of ICIs, ideal chemotherapy partners, and the financial implications of bringing newer therapies to the forefront. In this review, we discuss the evolving landscape of ICIs and ADCs in managing early-stage breast cancer and provide an overview of potential future advancement in the field.
Collapse
Affiliation(s)
- Prarthna V Bhardwaj
- Division of Hematology-Oncology, University of Massachusetts Chan School of Medicine-Baystate, Springfield, MA 01199, USA
| | - Yara G Abdou
- Division of Oncology, University of North Carolina at Chapel Hill, Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27514, USA
| |
Collapse
|
160
|
Hermida-Prado F, Xie Y, Sherman S, Nagy Z, Russo D, Akhshi T, Chu Z, Feit A, Campisi M, Chen M, Nardone A, Guarducci C, Lim K, Font-Tello A, Lee I, García-Pedrero J, Cañadas I, Agudo J, Huang Y, Sella T, Jin Q, Tayob N, Mittendorf EA, Tolaney SM, Qiu X, Long H, Symmans WF, Lin JR, Santagata S, Bedrosian I, Yardley DA, Mayer IA, Richardson ET, Oliveira G, Wu CJ, Schuster EF, Dowsett M, Welm AL, Barbie D, Metzger O, Jeselsohn R. Endocrine Therapy Synergizes with SMAC Mimetics to Potentiate Antigen Presentation and Tumor Regression in Hormone Receptor-Positive Breast Cancer. Cancer Res 2023; 83:3284-3304. [PMID: 37450351 PMCID: PMC10543960 DOI: 10.1158/0008-5472.can-23-1711] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
Immunotherapies have yet to demonstrate significant efficacy in the treatment of hormone receptor-positive (HR+) breast cancer. Given that endocrine therapy (ET) is the primary approach for treating HR+ breast cancer, we investigated the effects of ET on the tumor immune microenvironment (TME) in HR+ breast cancer. Spatial proteomics of primary HR+ breast cancer samples obtained at baseline and after ET from patients enrolled in a neoadjuvant clinical trial (NCT02764541) indicated that ET upregulated β2-microglobulin and influenced the TME in a manner that promotes enhanced immunogenicity. To gain a deeper understanding of the underlying mechanisms, the intrinsic effects of ET on cancer cells were explored, which revealed that ET plays a crucial role in facilitating the chromatin binding of RelA, a key component of the NF-κB complex. Consequently, heightened NF-κB signaling enhanced the response to interferon-gamma, leading to the upregulation of β2-microglobulin and other antigen presentation-related genes. Further, modulation of NF-κB signaling using a SMAC mimetic in conjunction with ET augmented T-cell migration and enhanced MHC-I-specific T-cell-mediated cytotoxicity. Remarkably, the combination of ET and SMAC mimetics, which also blocks prosurvival effects of NF-κB signaling through the degradation of inhibitors of apoptosis proteins, elicited tumor regression through cell autonomous mechanisms, providing additional support for their combined use in HR+ breast cancer. SIGNIFICANCE Adding SMAC mimetics to endocrine therapy enhances tumor regression in a cell autonomous manner while increasing tumor immunogenicity, indicating that this combination could be an effective treatment for HR+ patients with breast cancer.
Collapse
Affiliation(s)
- Francisco Hermida-Prado
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- University of Oviedo, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), IUOPA, Oviedo, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Yingtian Xie
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Shira Sherman
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Zsuzsanna Nagy
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Douglas Russo
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Tara Akhshi
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Zhengtao Chu
- Huntsman Cancer Institute, Department of Oncological Sciences, University of Utah, Salt Lake City, Utah
| | - Avery Feit
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Marco Campisi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Minyue Chen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Immunology, Harvard Medical School, Boston, Massachusetts
| | - Agostina Nardone
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Cristina Guarducci
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Klothilda Lim
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Alba Font-Tello
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Irene Lee
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Juana García-Pedrero
- University of Oviedo, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), IUOPA, Oviedo, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Israel Cañadas
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Judith Agudo
- Harvard Medical School, Boston, Massachusetts
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ying Huang
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Tal Sella
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, Massachusetts
| | - Qingchun Jin
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Nabihah Tayob
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Elizabeth A. Mittendorf
- Harvard Medical School, Boston, Massachusetts
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, Massachusetts
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Sara M. Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, Massachusetts
| | - Xintao Qiu
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Henry Long
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Jia-Ren Lin
- Ludwig Center at Harvard and Laboratory of Systems Pharmacology, Harvard Medical School, Boston, Massachusetts
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts
| | - Sandro Santagata
- Ludwig Center at Harvard and Laboratory of Systems Pharmacology, Harvard Medical School, Boston, Massachusetts
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Isabelle Bedrosian
- Department of Breast Surgical Oncology, Division of Surgery, MD Anderson Cancer Center, Houston, Texas
| | - Denise A. Yardley
- Department of Medical Oncology, Sarah Cannon Cancer Center, Nashville, Tennessee
- Tennessee Oncology, Nashville, Tennessee
| | - Ingrid A. Mayer
- Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee
| | - Edward T. Richardson
- Harvard Medical School, Boston, Massachusetts
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Giacomo Oliveira
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Catherine J. Wu
- Harvard Medical School, Boston, Massachusetts
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Eugene F. Schuster
- The BC Now Toby Robins Research Centre at the Institute of Cancer Research, London, United Kingdom
- Ralph Lauren Centre for BC Research, Royal Marsden Hospital, London, United Kingdom
- The Royal Marsden Hospital, London, United Kingdom
| | - Mitch Dowsett
- The BC Now Toby Robins Research Centre at the Institute of Cancer Research, London, United Kingdom
- Ralph Lauren Centre for BC Research, Royal Marsden Hospital, London, United Kingdom
- The Royal Marsden Hospital, London, United Kingdom
| | - Alana L. Welm
- Huntsman Cancer Institute, Department of Oncological Sciences, University of Utah, Salt Lake City, Utah
| | - David Barbie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Otto Metzger
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, Massachusetts
| | - Rinath Jeselsohn
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, Massachusetts
| |
Collapse
|
161
|
Kim L, Coman M, Pusztai L, Park TS. Neoadjuvant Immunotherapy in Early, Triple-Negative Breast Cancers: Catching Up with the Rest. Ann Surg Oncol 2023; 30:6441-6449. [PMID: 37349612 DOI: 10.1245/s10434-023-13714-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 05/23/2023] [Indexed: 06/24/2023]
Abstract
Despite breast cancer being long thought to be "immunologically cold," within early, triple-negative breast cancer (TNBC), there has been exciting advances with the use of immune checkpoint modulation combined with neoadjuvant chemotherapy. We review the major trials that have investigated combination immunochemotherapy in the neoadjuvant setting, reviewing both the pathological complete response rates and the maturing data regarding event-free and overall survival. Strategies to deescalate adjuvant therapy in patients with preserving excellent clinical outcome, as well as exploration of combinatorial adjuvant therapies to improve outcome in those with extensive residual are the next-generation challenges. In addition to refinement of existing biomarkers, such as PD-L1, TILs, and tumor mutational burden (TMB), exploration of topics like the microbiome as both a biomarker and a therapeutic has shown promise in other cancer types, which motivates investigating these in breast cancer.
Collapse
Affiliation(s)
- Leah Kim
- Section of Surgical Oncology, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Magdalena Coman
- Section of Surgical Oncology, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Lajos Pusztai
- Yale School of Medicine, Yale Cancer Center, New Haven, CT, USA
| | - Tristen S Park
- Section of Surgical Oncology, Department of Surgery, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
162
|
Zubareva E, Senchukova M, Karmakova T. Predictive significance of HIF-1α, Snail, and PD-L1 expression in breast cancer. Clin Exp Med 2023; 23:2369-2383. [PMID: 36802309 DOI: 10.1007/s10238-023-01026-z] [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: 01/11/2023] [Accepted: 02/08/2023] [Indexed: 02/23/2023]
Abstract
Currently, the prediction of breast cancer (BC) effectiveness to drug treatment is based on determining the expression level of steroid hormone receptors and human epidermal growth factor receptor type 2 (HER2). However, significant differences in individual response to drug treatment require the search for new predictive markers. Here, by comprehensively examining HIF-1α, Snail, and PD-L1 expression in BC tumor tissue, we demonstrate that high levels of these markers correlate with unfavorable factors of BC prognosis: the presence of regional and distant metastases and lymphovascular and perineural invasion. Analyzing the predictive significance of markers, we show that the most significant predictors of chemoresistant HER2-negative BC are a high PD-L1 level and a low Snail level, while in HER2-positive BC, only a high PD-L1 level is an independent predictor of chemoresistant BC. Our results suggest that using immune checkpoint inhibitors in these groups of patients may improve drug therapy effectiveness.
Collapse
Affiliation(s)
- Evgenia Zubareva
- Mammological Center, Orenburg Regional Clinical Oncology Center, Orenburg, Orenburg Region, Russian Federation, 460021
| | - Marina Senchukova
- Department of Oncology, Orenburg State Medical University, Orenburg, Orenburg Region, Russian Federation, 460000.
| | - Tatyana Karmakova
- Department of Predicting the Effectiveness of Conservative Therapy, P.A. Herzen Moscow Oncology Research Institute, Branch of the National Medical Research Radiological Center of the Ministry of Health of Russian Federation, Moscow, Moscow Region, Russian Federation, 125284
| |
Collapse
|
163
|
Downs-Canner S, Mittendorf EA. Selecting Triple Negative Breast Cancer Patients for Immunotherapy. Surg Oncol Clin N Am 2023; 32:733-745. [PMID: 37714640 DOI: 10.1016/j.soc.2023.05.005] [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] [Indexed: 09/17/2023]
Abstract
The approval of preoperative immunotherapy combined with chemotherapy is a practice-changing advance for patients with early-stage triple-negative breast cancer. The optimal patient selection requires careful attention to staging and balancing potential risks with expected benefits, particularly as it relates to immune-related adverse events.
Collapse
Affiliation(s)
- Stephanie Downs-Canner
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 300 East 66th Street, New York, NY 10065, USA. https://twitter.com/SDownsCanner
| | - Elizabeth A Mittendorf
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA; Breast Oncology Program, Dana-Farber Brigham Cancer Center, 450 Brookline Avenue, Boston, MA 02215, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
164
|
Cao H, Yao C, Yuan Y. Bayesian approach for design and analysis of medical device trials in the era of modern clinical studies. MEDICAL REVIEW (2021) 2023; 3:408-424. [PMID: 38283256 PMCID: PMC10810749 DOI: 10.1515/mr-2023-0026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 07/22/2023] [Indexed: 01/30/2024]
Abstract
Medical device technology develops rapidly, and the life cycle of a medical device is much shorter than drugs. It is necessary to evaluate the safety and effectiveness of a medical device in a timely manner to keep up with technology flux. Bayesian methods provides an efficient approach to addressing this challenge. In this article, we review the characteristics of the Bayesian approach and some Bayesian designs that were commonly used in medical device regulatory setting, including Bayesian adaptive design, Bayesian diagnostic design, Bayesian multiregional design, and Bayesian label expansion study. We illustrate these designs with medical devices approved by the US Food and Drug Administration (FDA). We also review several innovative Bayesian information borrowing methods, and briefly discuss the challenges and future directions of the Bayesian application in medical device trials. Our objective is to promote the use of the Bayesian approach to accelerate the development of innovative medical devices and their accessibility to patients for effective disease diagnoses and treatments.
Collapse
Affiliation(s)
- Han Cao
- Department of Biostatistics, Peking University First Hospital, Beijing, China
- Medical Data Science Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Chen Yao
- Department of Biostatistics, Peking University First Hospital, Beijing, China
- Peking University Clinical Research Institute, Beijing, China
- Hainan Institute of Real World Data, Qionghai, Hainan Province, China
| | - Ying Yuan
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, USA
| |
Collapse
|
165
|
Rustamadji P, Felicia D, Wuyung PE, Hellyanti T. The Role of Stromal Tumour Infiltrating Lymphocytes (sTIL) Intensity and Programmed Death Ligand 1 () Expression in Breast Cancer Response to Neoadjuvant Therapy in Cipto Mangunkusumo Hospital (CMH), Indonesia. Asian Pac J Cancer Prev 2023; 24:3459-3465. [PMID: 37898851 PMCID: PMC10770656 DOI: 10.31557/apjcp.2023.24.10.3459] [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: 04/13/2023] [Accepted: 10/19/2023] [Indexed: 10/30/2023] Open
Abstract
BACKGROUND Pathological responses to neoadjuvant therapy were still relatively poor, especially in CMH. Studies had been done to search for predictors of response such as sTIL intensity and expression, which is known to block sTIL action in killing cancer cells. This research assessed sTIL intensity and expression as predictors of response to neoadjuvant therapy in breast cancer. The preliminary data might be used to better tailored breast cancer patient therapy, considering the availability of anti-PD-1/ PD- L1 immunotherapy nowadays. OBJECTIVE To assess TIL intensity, expressions, and their roles as pathological predictors of breast cancer response to neoadjuvant therapy in Cipto Mangunkusumo Hospital (CMH). METHOD This was an observational analytic retrospective cohort study on breast cancer patients undergoing biopsy/review of biopsy specimens, receiving neoadjuvant therapy and mastectomy in CMH from January 2014 to December 2021. Sixty cases fulfilled the inclusion and exclusion criteria. Total sampling was done. expression (immunohistochemistry, clone 22C3) and sTIL intensity (histopathology) was examined in the biopsy specimen. Linear regression analysis was done to determine the independent predictors of neoadjuvant therapy response (evaluated in the mastectomy specimen with residual cancer burden/ RCB score). RESULTS There were 60 female patients, median age 46 years old. 91,7% had invasive carcinoma of no special type. Median sTIL intensity was 10% (1%-70%). 58,3% patients had low sTIL intensity (≤10%). 28,3% patients had positive expression (CPS ≥1). Only 8,3% patients had pCR, while 90% patients had RCB class II-III. Every 1% increase in sTIL intensity, no lymphovascular invasion, and taxane chemotherapy were predicted to lower RCB score by 0,058, 0,781, dan 0,594, respectively. expression associated with pCR-RCB class I (p=0,048), but CPS score was not a predictor of RCB score in linear regression analysis. CONSLUSION sTIL intensity was an independent predictor of breast cancer response to neoadjuvant therapy in RSCM. expression associated with pCR-RCB class I, but CPS score was not a predictor of RCB score.
Collapse
Affiliation(s)
| | - Devi Felicia
- Department of Anatomic Pathology, Faculty of Medicine Universitas Indonesia (FMUI)-Cipto Mangunkusumo Hospital (CMH), Jakarta, Indonesia.
| | | | | |
Collapse
|
166
|
Jin X, Zhou YF, Ma D, Zhao S, Lin CJ, Xiao Y, Fu T, Liu CL, Chen YY, Xiao WX, Liu YQ, Chen QW, Yu Y, Shi LM, Shi JX, Huang W, Robertson JFR, Jiang YZ, Shao ZM. Molecular classification of hormone receptor-positive HER2-negative breast cancer. Nat Genet 2023; 55:1696-1708. [PMID: 37770634 DOI: 10.1038/s41588-023-01507-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 08/21/2023] [Indexed: 09/30/2023]
Abstract
Hormone receptor-positive (HR+)/human epidermal growth factor receptor 2-negative (HER2-) breast cancer is the most prevalent type of breast cancer, in which endocrine therapy resistance and distant relapse remain unmet challenges. Accurate molecular classification is urgently required for guiding precision treatment. We established a large-scale multi-omics cohort of 579 patients with HR+/HER2- breast cancer and identified the following four molecular subtypes: canonical luminal, immunogenic, proliferative and receptor tyrosine kinase (RTK)-driven. Tumors of these four subtypes showed distinct biological and clinical features, suggesting subtype-specific therapeutic strategies. The RTK-driven subtype was characterized by the activation of the RTK pathways and associated with poor outcomes. The immunogenic subtype had enriched immune cells and could benefit from immune checkpoint therapy. In addition, we developed convolutional neural network models to discriminate these subtypes based on digital pathology for potential clinical translation. The molecular classification provides insights into molecular heterogeneity and highlights the potential for precision treatment of HR+/HER2- breast cancer.
Collapse
Affiliation(s)
- Xi Jin
- Key Laboratory of Breast Cancer, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yi-Fan Zhou
- Key Laboratory of Breast Cancer, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ding Ma
- Key Laboratory of Breast Cancer, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Shen Zhao
- Key Laboratory of Breast Cancer, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Cai-Jin Lin
- Key Laboratory of Breast Cancer, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yi Xiao
- Key Laboratory of Breast Cancer, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Tong Fu
- Key Laboratory of Breast Cancer, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Cheng-Lin Liu
- Key Laboratory of Breast Cancer, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yi-Yu Chen
- Key Laboratory of Breast Cancer, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Wen-Xuan Xiao
- Key Laboratory of Breast Cancer, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ya-Qing Liu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute and Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Qing-Wang Chen
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute and Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Ying Yu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute and Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Le-Ming Shi
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute and Shanghai Cancer Center, Fudan University, Shanghai, China
- International Human Phenome Institutes (Shanghai), Shanghai, China
| | - Jin-Xiu Shi
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies (SIBPT), Shanghai, China
| | - Wei Huang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies (SIBPT), Shanghai, China
| | | | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
| |
Collapse
|
167
|
Topalian SL, Forde PM, Emens LA, Yarchoan M, Smith KN, Pardoll DM. Neoadjuvant immune checkpoint blockade: A window of opportunity to advance cancer immunotherapy. Cancer Cell 2023; 41:1551-1566. [PMID: 37595586 PMCID: PMC10548441 DOI: 10.1016/j.ccell.2023.07.011] [Citation(s) in RCA: 131] [Impact Index Per Article: 65.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/09/2023] [Accepted: 07/27/2023] [Indexed: 08/20/2023]
Abstract
Among new treatment approaches for patients with cancer, few have accelerated as quickly as neoadjuvant immune checkpoint blockade (ICB). Neoadjuvant cancer therapy is administered before curative-intent surgery in treatment-naïve patients. Conventional neoadjuvant chemotherapy and radiotherapy are primarily intended to reduce tumor size, improving surgical resectability. However, recent scientific evidence outlined here suggests that neoadjuvant immunotherapy can expand and transcriptionally modify tumor-specific T cell clones to enhance both intratumoral and systemic anti-tumor immunity. It further offers a unique "window of opportunity" to explore mechanisms and identify novel biomarkers of ICB response and resistance, opening possibilities for refining long-term clinical outcome predictions and developing new, more highly effective ICB combination therapies. Here, we examine advances in clinical and scientific knowledge gleaned from studies in select cancers and describe emerging key principles relevant to neoadjuvant ICB across many cancer types.
Collapse
Affiliation(s)
- Suzanne L Topalian
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Bloomberg-Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | - Patrick M Forde
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Bloomberg-Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | - Mark Yarchoan
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Bloomberg-Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Kellie N Smith
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Bloomberg-Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Drew M Pardoll
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Bloomberg-Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
168
|
Liu Y, Hu Y, Xue J, Li J, Yi J, Bu J, Zhang Z, Qiu P, Gu X. Advances in immunotherapy for triple-negative breast cancer. Mol Cancer 2023; 22:145. [PMID: 37660039 PMCID: PMC10474743 DOI: 10.1186/s12943-023-01850-7] [Citation(s) in RCA: 107] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 08/26/2023] [Indexed: 09/04/2023] Open
Abstract
BACKGROUND Immunotherapy has recently emerged as a treatment strategy which stimulates the human immune system to kill tumor cells. Tumor immunotherapy is based on immune editing, which enhances the antigenicity of tumor cells and increases the tumoricidal effect of immune cells. It also suppresses immunosuppressive molecules, activates or restores immune system function, enhances anti-tumor immune responses, and inhibits the growth f tumor cell. This offers the possibility of reducing mortality in triple-negative breast cancer (TNBC). MAIN BODY Immunotherapy approaches for TNBC have been diversified in recent years, with breakthroughs in the treatment of this entity. Research on immune checkpoint inhibitors (ICIs) has made it possible to identify different molecular subtypes and formulate individualized immunotherapy schedules. This review highlights the unique tumor microenvironment of TNBC and integrates and analyzes the advances in ICI therapy. It also discusses strategies for the combination of ICIs with chemotherapy, radiation therapy, targeted therapy, and emerging treatment methods such as nanotechnology, ribonucleic acid vaccines, and gene therapy. Currently, numerous ongoing or completed clinical trials are exploring the utilization of immunotherapy in conjunction with existing treatment modalities for TNBC. The objective of these investigations is to assess the effectiveness of various combined immunotherapy approaches and determine the most effective treatment regimens for patients with TNBC. CONCLUSION This review provides insights into the approaches used to overcome drug resistance in immunotherapy, and explores the directions of immunotherapy development in the treatment of TNBC.
Collapse
Affiliation(s)
- Yang Liu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Yueting Hu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Jinqi Xue
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Jingying Li
- Department of Health Management, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Jiang Yi
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Jiawen Bu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Zhenyong Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China.
| | - Peng Qiu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China.
| | - Xi Gu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China.
| |
Collapse
|
169
|
Huppert LA, Gumusay O, Idossa D, Rugo HS. Systemic therapy for hormone receptor-positive/human epidermal growth factor receptor 2-negative early stage and metastatic breast cancer. CA Cancer J Clin 2023; 73:480-515. [PMID: 36939293 DOI: 10.3322/caac.21777] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 01/09/2023] [Accepted: 02/01/2023] [Indexed: 03/21/2023] Open
Abstract
Hormone receptor (HR)-positive and human epidermal growth factor receptor 2 (HER2)-negative breast cancer is defined by the presence of the estrogen receptor and/or the progesterone receptor and the absence of HER2 gene amplification. HR-positive/HER2-negative breast cancer accounts for 65%-70% of all breast cancers, and incidence increases with increasing age. Treatment varies by stage, and endocrine therapy is the mainstay of treatment in both early stage and late-stage disease. Combinations with cyclin-dependent kinase 4/6 inhibitors have reduced distant recurrence in the early stage setting and improved overall survival in the metastatic setting. Chemotherapy is used based on stage and tumor biology in the early stage setting and after endocrine resistance for advanced disease. New therapies, including novel endocrine agents and antibody-drug conjugates, are now changing the treatment landscape. With the availability of new treatment options, it is important to define the optimal sequence of treatment to maximize clinical benefit while minimizing toxicity. In this review, the authors first discuss the pathologic and molecular features of HR-positive/HER2-negative breast cancer and mechanisms of endocrine resistance. Then, they discuss current and emerging therapies for both early stage and metastatic HR-positive/HER2-negative breast cancer, including treatment algorithms based on current data.
Collapse
Affiliation(s)
- Laura A Huppert
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, California, USA
| | - Ozge Gumusay
- Department of Medical Oncology, Acibadem University, School of Medicine, Istanbul, Turkey
| | - Dame Idossa
- Masonic Comprehensive Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Hope S Rugo
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, California, USA
| |
Collapse
|
170
|
Jiang S, Liu Y, Zheng H, Zhang L, Zhao H, Sang X, Xu Y, Lu X. Evolutionary patterns and research frontiers in neoadjuvant immunotherapy: a bibliometric analysis. Int J Surg 2023; 109:2774-2783. [PMID: 37216225 PMCID: PMC10498839 DOI: 10.1097/js9.0000000000000492] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 05/08/2023] [Indexed: 05/24/2023]
Abstract
Research has shown that neoadjuvant immunotherapy may provide more significant clinical benefits to cancer patients undergoing surgery than adjuvant therapy. This study examines the development of neoadjuvant immunotherapy research using bibliometric analysis. As of 12 February 2023, articles on neoadjuvant immunotherapy in the Web of Science Core Collection were collected. Co-authorship and keyword co-occurrence analyses and visualizations were performed using VOSviewer, while CiteSpace was used to identify bursting keywords and references. The study analyzed a total of 1222 neoadjuvant immunotherapy publications. The top contributors to this field were the United States, China, and Italy, and the journal with the most publications was Frontiers in Oncology. Francesco Montorsi had the highest H-index. The most common keywords were 'immunotherapy' and 'neoadjuvant therapy'. The study conducted a bibliometric analysis of over 20 years of neoadjuvant immunotherapy research, identifying the countries, institutions, authors, journals, and publications involved in this field. The findings provide a comprehensive overview of neoadjuvant immunotherapy research.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yiyao Xu
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Lu
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
171
|
Li J, Shen G, Wang M, Huo X, Zhao F, Ren D, Zhao Y, Zhao J. Comparative efficacy and safety of first-line neoadjuvant treatments in triple-negative breast cancer: systematic review and network meta-analysis. Clin Exp Med 2023; 23:1489-1499. [PMID: 36152119 DOI: 10.1007/s10238-022-00894-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 09/12/2022] [Indexed: 11/30/2022]
Abstract
Neoadjuvant treatment options for triple-negative breast cancer (TNBC) are abundant, but the efficacy of different combinations of treatment options remains unclear. Our network meta-analysis aimed to evaluate the effectiveness and safety of various neoadjuvant treatment options in patients with TNBC. Literature reports published before March 31, 2022, were retrieved from the PubMed, Embase, Cochrane Library, main oncology conference of the European Society of Medical Oncology, American Society of Clinical Oncology, and San Antonio Breast Cancer Symposium databases. Pairwise and Bayesian network meta-analyses were performed to compare direct and indirect evidence, respectively. The primary outcome was pathological complete response (pCR). Comparison of efficiency between different treatment regimens was made by HRs and 95% confidence intervals (CIs). Overall, 26 studies, including 9714 TNBC patients, were assessed in this network meta-analysis. Results indicated that the pCR of immune checkpoint inhibitors plus platinum-containing regimens is better than other joint regimens. PCR rate of neoadjuvant chemotherapy regimens containing bevacizumab, platinum, poly(ADP-ribose) polymerase inhibitors, and immune checkpoint inhibitors was higher than those of standard chemotherapy agents. By performing a conjoint analysis of the pCR rate and safety endpoints, we found that immune checkpoint inhibitors plus platinum-containing regimens were well balanced in terms of efficacy and toxicity. Considering the efficacy and acceptable adverse events, neoadjuvant chemotherapy based on immune checkpoint inhibitors plus platinum may be considered as an option for patients with TNBC.
Collapse
Affiliation(s)
- Jinming Li
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Guoshuang Shen
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Miaozhou Wang
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Xingfa Huo
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Fuxing Zhao
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Dengfeng Ren
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Yi Zhao
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China.
| | - Jiuda Zhao
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China.
| |
Collapse
|
172
|
Abaza A, Sid Idris F, Anis Shaikh H, Vahora I, Moparthi KP, Al Rushaidi MT, Muddam MR, Obajeun OA, Jaramillo AP, Khan S. Programmed Cell Death Protein 1 (PD-1) and Programmed Cell Death Ligand 1 (PD-L1) Immunotherapy: A Promising Breakthrough in Cancer Therapeutics. Cureus 2023; 15:e44582. [PMID: 37667784 PMCID: PMC10475160 DOI: 10.7759/cureus.44582] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 09/02/2023] [Indexed: 09/06/2023] Open
Abstract
The advent of immune checkpoint inhibitors has revolutionized cancer therapy by leveraging the body's immune system to combat malignancies effectively. Among these groundbreaking agents, programmed cell death protein 1 (PD-1) and programmed cell death ligand 1 (PD-L1) inhibitors have emerged as pivotal therapeutic approaches. PD-L1, a key protein expressed on the surface of various cells, including cancer cells, plays a central role in immune regulation by interacting with the programmed cell death protein 1 (PD-1) receptor on T-cells leading to immune suppression. The substantial increase in PD-L1 expression on cancer cell surfaces has driven the exploration of PD-1/PD-L1 inhibitors as potential immunotherapeutic agents. These inhibitors are monoclonal antibodies designed to impede the PD-L1 and PD-1 interaction and disrupt the immunosuppressive signal, thereby reinvigorating the anti-tumor immune response mediated by activated T-cells. Clinical trials investigating PD-1/PD-L1 inhibitors have demonstrated remarkable efficacy in the treatment of diverse advanced or metastatic cancers, including leukemia, non-small cell lung (NSCLC), hepatocellular, melanoma, gastric, colorectal, and breast cancers, among others. Regulatory approvals have been granted for both monotherapy and combination therapy with other cancer treatments, encompassing chemotherapy and additional immune checkpoint inhibitors. While PD-1/PD-L1 inhibitors have exhibited significant success, they are not devoid of challenges. The emergence of intrinsic or acquired resistance, as well as immune-related adverse events, warrants thorough investigation and management. Consequently, researchers have embarked on combination trials to augment the therapeutic potential of PD-1/PD-L1 inhibitors and surmount resistance mechanisms.
Collapse
Affiliation(s)
- Abdelrahman Abaza
- Pathology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Faten Sid Idris
- Pediatrics, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Humna Anis Shaikh
- Pediatrics, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Ilma Vahora
- General Surgery, Saint George's University School of Medicine, Chicago, USA
| | - Kiran Prasad Moparthi
- College of Medicine, Sri Venkata Sai (SVS) Medical College, Mahabubnagar, IND
- General Practice, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Majdah T Al Rushaidi
- Psychology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Meghana Reddy Muddam
- General Practice, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- College of Medicine, Sri Venkata Sai (SVS) Medical College, Mahabubnagar, IND
| | - Omobolanle A Obajeun
- Paediatrics, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | | | - Safeera Khan
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
173
|
Zhao X, Dong F, Chen W, Liu C. Immunotherapy in combination with neoadjuvant anthracycline‑free chemotherapy for triple‑negative early breast cancer: A case report. Oncol Lett 2023; 26:374. [PMID: 37559578 PMCID: PMC10407723 DOI: 10.3892/ol.2023.13961] [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: 02/18/2023] [Accepted: 06/01/2023] [Indexed: 08/11/2023] Open
Abstract
Immunotherapy is a promising anticancer strategy. In the present report, the case of a 36-year-old female patient with pathologically diagnosed, left triple-negative breast cancer and axillary lymph node involvement is reported. The patient received immunotherapy in combination with neoadjuvant anthracycline-free chemotherapy for six cycles, before undergoing left mastectomy and left axillary lymph node dissection. The postoperative pathology was a complete response to treatment, involving eradication of tumor from both the breast and the relevant lymph nodes. However, thyroid dysfunction occurred after two cycles of neoadjuvant treatment. The clinical presentation of the thyroid disorder was transient hyperthyroidism for 4 weeks and subsequent hypothyroidism, which required hormone replacement therapy.
Collapse
Affiliation(s)
- Xiangwang Zhao
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Fang Dong
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Weibin Chen
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Chunping Liu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
174
|
Laouris P, Rodrigues MES, Costa SCV, Rala de Paula BH. Neoadjuvant immunotherapy in early-stage triple negative breast cancer. ANNALS OF TRANSLATIONAL MEDICINE 2023; 11:338. [PMID: 37675326 PMCID: PMC10477652 DOI: 10.21037/atm-23-1550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 05/30/2023] [Indexed: 09/08/2023]
Affiliation(s)
- Panayiotis Laouris
- Early Phase Trials Unit, Division of Oncology, University of Cambridge, Cambridge, UK
| | | | | | | |
Collapse
|
175
|
Zhu S, Wu Y, Song B, Yi M, Yan Y, Mei Q, Wu K. Recent advances in targeted strategies for triple-negative breast cancer. J Hematol Oncol 2023; 16:100. [PMID: 37641116 PMCID: PMC10464091 DOI: 10.1186/s13045-023-01497-3] [Citation(s) in RCA: 109] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/21/2023] [Indexed: 08/31/2023] Open
Abstract
Triple-negative breast cancer (TNBC), a highly aggressive subtype of breast cancer, negatively expresses estrogen receptor, progesterone receptor, and the human epidermal growth factor receptor 2 (HER2). Although chemotherapy is the main form of treatment for patients with TNBC, the effectiveness of chemotherapy for TNBC is still limited. The search for more effective therapies is urgent. Multiple targeted therapeutic strategies have emerged according to the specific molecules and signaling pathways expressed in TNBC. These include PI3K/AKT/mTOR inhibitors, epidermal growth factor receptor inhibitors, Notch inhibitors, poly ADP-ribose polymerase inhibitors, and antibody-drug conjugates. Moreover, immune checkpoint inhibitors, for example, pembrolizumab, atezolizumab, and durvalumab, are widely explored in the clinic. We summarize recent advances in targeted therapy and immunotherapy in TNBC, with the aim of serving as a reference for the development of individualized treatment of patients with TNBC in the future.
Collapse
Affiliation(s)
- Shuangli Zhu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuze Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Bin Song
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| | - Ming Yi
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Yuheng Yan
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qi Mei
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China.
- Cancer Center, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Kongming Wu
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China.
- Cancer Center, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
176
|
Connell W, Garcia K, Goodarzi H, Keiser MJ. Learning chemical sensitivity reveals mechanisms of cellular response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.26.554851. [PMID: 37693536 PMCID: PMC10491110 DOI: 10.1101/2023.08.26.554851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Chemical probes interrogate disease mechanisms at the molecular level by linking genetic changes to observable traits. However, comprehensive chemical screens in diverse biological models are impractical. To address this challenge, we developed ChemProbe, a model that predicts cellular sensitivity to hundreds of molecular probes and drugs by learning to combine transcriptomes and chemical structures. Using ChemProbe, we inferred the chemical sensitivity of cancer cell lines and tumor samples and analyzed how the model makes predictions. We retrospectively evaluated drug response predictions for precision breast cancer treatment and prospectively validated chemical sensitivity predictions in new cellular models, including a genetically modified cell line. Our model interpretation analysis identified transcriptome features reflecting compound targets and protein network modules, identifying genes that drive ferroptosis. ChemProbe is an interpretable in silico screening tool that allows researchers to measure cellular response to diverse compounds, facilitating research into molecular mechanisms of chemical sensitivity.
Collapse
Affiliation(s)
- William Connell
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Kristle Garcia
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Hani Goodarzi
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Michael J. Keiser
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
177
|
Bhardwaj PV, Wang Y, Brunk E, Spanheimer PM, Abdou YG. Advances in the Management of Early-Stage Triple-Negative Breast Cancer. Int J Mol Sci 2023; 24:12478. [PMID: 37569851 PMCID: PMC10419523 DOI: 10.3390/ijms241512478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer with both inter- and intratumor heterogeneity, thought to result in a more aggressive course and worse outcomes. Neoadjuvant therapy (NAT) has become the preferred treatment modality of early-stage TNBC as it allows for the downstaging of tumors in the breast and axilla, monitoring early treatment response, and most importantly, provides important prognostic information that is essential to determining post-surgical therapies to improve outcomes. It focuses on combinations of systemic drugs to optimize pathologic complete response (pCR). Excellent response to NAT has allowed surgical de-escalation in ideal candidates. Further, treatment algorithms guide the systemic management of patients based on their pCR status following surgery. The expanding knowledge of molecular pathways, genomic sequencing, and the immunological profile of TNBC has led to the use of immune checkpoint inhibitors and targeted agents, including PARP inhibitors, further revolutionizing the therapeutic landscape of this clinical entity. However, subgroups most likely to benefit from these novel approaches in TNBC remain elusive and are being extensively studied. In this review, we describe current practices and promising therapeutic options on the horizon for TNBC, surgical advances, and future trends in molecular determinants of response to therapy in early-stage TNBC.
Collapse
Affiliation(s)
- Prarthna V. Bhardwaj
- Division of Hematology-Oncology, University of Massachusetts Chan Medical School—Baystate, Springfield, MA 01199, USA
| | - Yue Wang
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Curriculum in Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Elizabeth Brunk
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Integrative Program for Biological and Genomic Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, UNC Chapel Hill, NC 27599, USA
- Computational Medicine Program, UNC Chapel Hill, NC 27599, USA
| | - Philip M. Spanheimer
- Lineberger Comprehensive Cancer Center, UNC Chapel Hill, NC 27599, USA
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yara G. Abdou
- Lineberger Comprehensive Cancer Center, UNC Chapel Hill, NC 27599, USA
- Division of Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
178
|
Nguyen VP, Campbell KM, Nowicki TS, Elumalai N, Medina E, Baselga-Carretero I, DiNome ML, Chang HR, Oseguera DK, Ribas A, Glaspy JA. A Pilot Study of Neoadjuvant Nivolumab, Ipilimumab, and Intralesional Oncolytic Virotherapy for HER2-negative Breast Cancer. CANCER RESEARCH COMMUNICATIONS 2023; 3:1628-1637. [PMID: 37621406 PMCID: PMC10445661 DOI: 10.1158/2767-9764.crc-23-0145] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 08/26/2023]
Abstract
Purpose Neoadjuvant combination immune checkpoint blockade and intralesional oncolytic virotherapy have the potential to activate antitumor responses in patients with breast cancer. Experimental Design Eligibility for this pilot phase I trial included patients with localized HER2-negative breast cancer who received systemic nivolumab and ipilimumab and intratumor talimogene laherparepvec (T-VEC; NCT04185311). The primary objective was to evaluate the safety and adverse event profile of immunotherapy combined with T-VEC in patients with localized, HER2-negative breast cancer. Results Six patients were enrolled, 4 having relapses after prior neoadjuvant chemotherapy and 2 who were previously untreated. Toxicities included 1 patient having grade 3 hypotension and type 1 diabetes mellitus, 3 patients with hypothyroidism, and all patients having constitutional symptoms known to be associated with the administration of T-VEC. One patient had a pathologic complete response, 3 patients had pathologic partial responses, 1 showed no significant response, and 1 had disease progression. Biopsies demonstrated increased immune cell infiltration in samples from patients who responded to therapy. Conclusions This triple immunotherapy regimen provided responses in patients with advanced or relapsed HER2-negative breast cancer, at the expense of long-term toxicities. Significance Systemic immune checkpoint blockade with a programmed death receptor 1 and a CTL antigen-4 blocking antibody, combined with intralesional oncolytic virotherapy, is a chemotherapy-free combination aimed at inducing an antitumor immune response locally and systemic immunity.
Collapse
Affiliation(s)
- Vina P. Nguyen
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles, Los Angeles, California
| | - Katie M. Campbell
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California
| | - Theodore S. Nowicki
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California
- Parker Institute for Cancer Immunotherapy, San Francisco, California
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of California, Los Angeles, Los Angeles, California
- Department of Microbiology, Immunology and Genetics, University of California, Los Angeles, Los Angeles, California
| | - Nila Elumalai
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles, Los Angeles, California
| | - Egmidio Medina
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles, Los Angeles, California
| | - Ignacio Baselga-Carretero
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles, Los Angeles, California
| | - Maggie L. DiNome
- Department of Surgery, Division of Surgical Oncology, University of California, Los Angeles, Los Angeles, California
| | - Helena R. Chang
- Department of Surgery, Division of Surgical Oncology, University of California, Los Angeles, Los Angeles, California
| | - Denise K. Oseguera
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles, Los Angeles, California
| | - Antoni Ribas
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California
- Department of Microbiology, Immunology and Genetics, University of California, Los Angeles, Los Angeles, California
- Department of Surgery, Division of Surgical Oncology, University of California, Los Angeles, Los Angeles, California
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California
| | - John A. Glaspy
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
179
|
Jahandideh A, Yarizadeh M, Noei-Khesht Masjedi M, Fatehnejad M, Jahandideh R, Soheili R, Eslami Y, Zokaei M, Ahmadvand A, Ghalamkarpour N, Kumar Pandey R, Nabi Afjadi M, Payandeh Z. Macrophage's role in solid tumors: two edges of a sword. Cancer Cell Int 2023; 23:150. [PMID: 37525217 PMCID: PMC10391843 DOI: 10.1186/s12935-023-02999-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 07/24/2023] [Indexed: 08/02/2023] Open
Abstract
The tumor microenvironment is overwhelmingly dictated by macrophages, intimately affiliated with tumors, exercising pivotal roles in multiple processes, including angiogenesis, extracellular matrix reconfiguration, cellular proliferation, metastasis, and immunosuppression. They further exhibit resilience to chemotherapy and immunotherapy via meticulous checkpoint blockades. When appropriately stimulated, macrophages can morph into a potent bidirectional component of the immune system, engulfing malignant cells and annihilating them with cytotoxic substances, thus rendering them intriguing candidates for therapeutic targets. As myelomonocytic cells relentlessly amass within tumor tissues, macrophages rise as prime contenders for cell therapy upon the development of chimeric antigen receptor effector cells. Given the significant incidence of macrophage infiltration correlated with an unfavorable prognosis and heightened resistance to chemotherapy in solid tumors, we delve into the intricate role of macrophages in cancer propagation and their promising potential in confronting four formidable cancer variants-namely, melanoma, colon, glioma, and breast cancers.
Collapse
Affiliation(s)
- Arian Jahandideh
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
- Usern Office, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mahsa Yarizadeh
- Islamic Azad University, Tehran Medical Branch, Tehran, Iran
| | - Maryam Noei-Khesht Masjedi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Fatehnejad
- Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Romina Jahandideh
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Roben Soheili
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Science, Islamic Azad University, Tehran, Iran
| | - Yeganeh Eslami
- Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Maryam Zokaei
- Department of Food Science and Technology, Faculty of Nutrition Science, Food Science and Technology/National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ardavan Ahmadvand
- Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nogol Ghalamkarpour
- Department of Clinical Laboratory Sciences, School of Allied Medicine, Student Research Committee, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Rajan Kumar Pandey
- Department Medical Biochemistry and Biophysics, Division Medical Inflammation Research, Karolinska Institute, Stockholm, Sweden
| | - Mohsen Nabi Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Zahra Payandeh
- Department Medical Biochemistry and Biophysics, Division Medical Inflammation Research, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
180
|
Moyer CL, Brown PH. Targeting nuclear hormone receptors for the prevention of breast cancer. Front Med (Lausanne) 2023; 10:1200947. [PMID: 37583424 PMCID: PMC10424511 DOI: 10.3389/fmed.2023.1200947] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/30/2023] [Indexed: 08/17/2023] Open
Abstract
Advancements in research have led to the steady decline of breast cancer mortality over the past thirty years. However, breast cancer incidence has continued to rise, resulting in an undue burden on healthcare costs and highlighting a great need for more effective breast cancer prevention strategies, including targeted chemo preventative agents. Efforts to understand the etiology of breast cancer have uncovered important roles for nuclear receptors in the development and progression of breast cancer. Targeted therapies to inhibit estrogen receptor (ER) and progesterone receptor (PR) signaling (selective ER modulators, aromatase inhibitors and selective PR modulators) have shown great promise for the treatment and prevention of hormone receptor (HR)-positive breast cancer. However, these drugs do not prevent HR-negative disease. Therefore, recent efforts have focused on novel targeted therapies with the potential to prevent both HR-positive and HR-negative breast cancer. Among these include drugs that target other nuclear receptors, such as retinoic acid receptor (RAR), retinoid X receptor (RXR) and vitamin D receptor (VDR). In this review we provide an overview of recent preclinical and clinical trials targeting members of the nuclear receptor superfamily for the prevention of breast cancer.
Collapse
Affiliation(s)
- Cassandra L. Moyer
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Powel H. Brown
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
181
|
Boydell E, Sandoval JL, Michielin O, Obeid M, Addeo A, Friedlaender A. Neoadjuvant Immunotherapy: A Promising New Standard of Care. Int J Mol Sci 2023; 24:11849. [PMID: 37511609 PMCID: PMC10380420 DOI: 10.3390/ijms241411849] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
Neoadjuvant immunotherapy has emerged as a promising approach in the treatment of various malignancies, with preclinical studies showing improved immune responses in the preoperative setting. FDA-approved neoadjuvant-immunotherapy-based approaches include triple-negative breast cancer and early non-small cell lung cancer on the basis of improvement in pathological response and event free survival. Nevertheless, current trials have only shown benefits in a fraction of patients. It is therefore crucial to identify predictive biomarkers to improve patient selection for such approaches. This review aims to provide an overview of potential biomarkers of neoadjuvant immunotherapy in early triple-negative breast cancer, bladder cancer, melanoma, non-small cell lung cancer, colorectal cancer and gastric cancer. By the extrapolation of the metastatic setting, we explore known predictive biomarkers, i.e., PD-L1, mismatch repair deficiency and tumour mutational burden, as well as potential early-disease-specific biomarkers. We also discuss the challenges of identifying reliable biomarkers and the need for standardized protocols and guidelines for their validation and clinical implementation.
Collapse
Affiliation(s)
- Emma Boydell
- University Hospital of Geneva, 1205 Geneva, Switzerland
| | | | | | - Michel Obeid
- University Hospital of Lausanne, 1005 Lausanne, Switzerland
| | - Alfredo Addeo
- University Hospital of Geneva, 1205 Geneva, Switzerland
| | - Alex Friedlaender
- University Hospital of Geneva, 1205 Geneva, Switzerland
- Clinique Générale Beaulieu, 1206 Geneva, Switzerland
| |
Collapse
|
182
|
Qian D, Xu Y, Wu Y, Qiu J, Hong W, Meng X. Assessment of the safety and efficacy of combination chemotherapy and PD-1/PD-L1 inhibitor treatment of breast cancer: A meta-analysis. Chin Med J (Engl) 2023; 136:1663-1670. [PMID: 37279392 PMCID: PMC10344516 DOI: 10.1097/cm9.0000000000002631] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Indexed: 06/08/2023] Open
Abstract
BACKGROUND As the efficacy of programmed cell death-1/programmed death-ligand 1 (PD-1/PD-L1) inhibitors combined with chemotherapy in curing breast cancer is still controversial, this meta-analysis compares the efficacy and safety of PD-1/PD-L1 inhibitors combined with chemotherapy and chemotherapy alone in the treatment of breast cancer, which provides guidance for the clinical treatment. METHODS Relevant studies published as of April 2022 in the various databases including EMBASE, PubMed, and Cochrane Library were selected. Randomized controlled trials (RCTs) in which control patients underwent chemotherapy alone and experimental group patients underwent combination chemotherapy and PD-1/PD-L1 inhibitor treatment were included in this investigation. Investigations without complete information, researches from which information could not be extracted, duplicate articles, animal studies, review articles, and systematic reviews were excluded. STATA 15.1 was employed for all statistical analyses. RESULTS In total, eight eligible studies were identified, revealing that combination chemotherapy and PD-1/PD-L1 inhibitor treatment was linked to significant increases in progression-free survival (PFS) relative to chemotherapy alone (hazard ratio [HR] = 0.83, 95% confidence interval [CI]: 0.70-0.99, P = 0.032) but not overall survival (HR = 0.92, 95% CI: 0.80-1.06, P = 0.273). Pooled adverse event rates were also increased within the group of combination treatment relative to the chemotherapy group (risk ratio [RR] = 1.08, 95% CI: 1.03-1.14, P = 0.002). Specifically, nausea rates were lesser within the group of combination treatment relative to the group of chemotherapy (RR = 0.48, 95% CI: 0.25-0.92, P = 0.026). Subgroup analyses indicated that the PFS of patients who underwent combination atezolizumab or pembrolizumab and chemotherapy treatment were substantially longer than those of patients who underwent chemotherapy alone (HR = 0.79, 95% CI: 0.69-0.89, P ≤0.001; HR = 0.79, 95% CI: 0.67-0.92, P = 0.002). CONCLUSIONS The pooled results suggest that combination chemotherapy and PD-1/PD-L1 inhibitor treatment approaches help prolong PFS in breast cancer patients, but have no statistically significant effect on overall survival (OS). Additionally, combination therapy can significantly improve complete response rate (CRR) compared with chemotherapy alone. However, combination therapy was associated with greater rates of adverse events.
Collapse
Affiliation(s)
- Da Qian
- Department of Burn and Plastic Surgery-Hand Surgery, The Changshu Hospital Affiliated to Soochow University, Changshu, Soochow, Jiangsu 215500, China
| | - Yuhao Xu
- Second Clinical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Yihao Wu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, Zhejiang 310000, China
| | - Jie Qiu
- Second Clinical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Weimin Hong
- Faculty of Basic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310000, China
| | - Xuli Meng
- Department of Breast Surgery, Cancer Center, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310000, China
| |
Collapse
|
183
|
Srivastava N, Usmani SS, Subbarayan R, Saini R, Pandey PK. Hypoxia: syndicating triple negative breast cancer against various therapeutic regimens. Front Oncol 2023; 13:1199105. [PMID: 37492478 PMCID: PMC10363988 DOI: 10.3389/fonc.2023.1199105] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 06/05/2023] [Indexed: 07/27/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is one of the deadliest subtypes of breast cancer (BC) for its high aggressiveness, heterogeneity, and hypoxic nature. Based on biological and clinical observations the TNBC related mortality is very high worldwide. Emerging studies have clearly demonstrated that hypoxia regulates the critical metabolic, developmental, and survival pathways in TNBC, which include glycolysis and angiogenesis. Alterations to these pathways accelerate the cancer stem cells (CSCs) enrichment and immune escape, which further lead to tumor invasion, migration, and metastasis. Beside this, hypoxia also manipulates the epigenetic plasticity and DNA damage response (DDR) to syndicate TNBC survival and its progression. Hypoxia fundamentally creates the low oxygen condition responsible for the alteration in Hypoxia-Inducible Factor-1alpha (HIF-1α) signaling within the tumor microenvironment, allowing tumors to survive and making them resistant to various therapies. Therefore, there is an urgent need for society to establish target-based therapies that overcome the resistance and limitations of the current treatment plan for TNBC. In this review article, we have thoroughly discussed the plausible significance of HIF-1α as a target in various therapeutic regimens such as chemotherapy, radiotherapy, immunotherapy, anti-angiogenic therapy, adjuvant therapy photodynamic therapy, adoptive cell therapy, combination therapies, antibody drug conjugates and cancer vaccines. Further, we also reviewed here the intrinsic mechanism and existing issues in targeting HIF-1α while improvising the current therapeutic strategies. This review highlights and discusses the future perspectives and the major alternatives to overcome TNBC resistance by targeting hypoxia-induced signaling.
Collapse
Affiliation(s)
- Nityanand Srivastava
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Salman Sadullah Usmani
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Rajasekaran Subbarayan
- Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY, United States
- Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Educations, Chennai, India
| | - Rashmi Saini
- Department of Zoology, Gargi College, University of Delhi, New Delhi, India
| | - Pranav Kumar Pandey
- Dr. R.P. Centre for Opthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
184
|
Nederlof I, Voorwerk L, Kok M. Facts and Hopes in Immunotherapy for Early-Stage Triple-Negative Breast Cancer. Clin Cancer Res 2023; 29:2362-2370. [PMID: 36622327 PMCID: PMC10320476 DOI: 10.1158/1078-0432.ccr-22-0701] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/24/2022] [Accepted: 12/02/2022] [Indexed: 01/10/2023]
Abstract
A substantial fraction of early-stage triple-negative breast cancer (eTNBC) is characterized by high levels of stromal tumor-infiltrating lymphocytes (sTIL) and has a good prognosis even without systemic treatment, highlighting the importance of an endogenous anticancer immune response. Still, a considerable proportion of patients with eTNBC need some "therapeutical push" to kick-start this immune response. Exploiting this immune response with immune-checkpoint inhibition (ICI), in combination with chemotherapy, has made its way into standard of care in eTNBC. Major challenges in the near future include finding those patients with eTNBC who can be treated with ICI alone or with a reduced chemotherapy backbone. Exploring the optimal duration of ICI and finding biomarkers to predict response will be key to enable personalized implementation of ICI in patients with eTNBC. For patients who currently do not respond effectively to ICI plus chemotherapy, challenges lie in finding new immunomodulatory therapies and developing response-guided neoadjuvant approaches.
Collapse
Affiliation(s)
- Iris Nederlof
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Leonie Voorwerk
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Marleen Kok
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
185
|
Aristei C, Kaidar-Person O, Boersma L, Leonardi MC, Offersen B, Franco P, Arenas M, Bourgier C, Pfeffer R, Kouloulias V, Bölükbaşı Y, Meattini I, Coles C, Luis AM, Masiello V, Palumbo I, Morganti AG, Perrucci E, Tombolini V, Krengli M, Marazzi F, Trigo L, Borghesi S, Ciabattoni A, Ratoša I, Valentini V, Poortmans P. The 2022 Assisi Think Tank Meeting: White paper on optimising radiation therapy for breast cancer. Crit Rev Oncol Hematol 2023; 187:104035. [PMID: 37244324 DOI: 10.1016/j.critrevonc.2023.104035] [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: 02/23/2023] [Revised: 05/11/2023] [Accepted: 05/23/2023] [Indexed: 05/29/2023] Open
Abstract
The present white paper, referring to the 4th Assisi Think Tank Meeting on breast cancer, reviews state-of-the-art data, on-going studies and research proposals. <70% agreement in an online questionnaire identified the following clinical challenges: 1: Nodal RT in patients who have a) 1-2 positive sentinel nodes without ALND (axillary lymph node dissection); b) cN1 disease transformed into ypN0 by primary systemic therapy and c) 1-3 positive nodes after mastectomy and ALND. 2. The optimal combination of RT and immunotherapy (IT), patient selection, IT-RT timing, and RT optimal dose, fractionation and target volume. Most experts agreed that RT- IT combination does not enhance toxicity. 3: Re-irradiation for local relapse converged on the use of partial breast irradiation after second breast conserving surgery. Hyperthermia aroused support but is not widely available. Further studies are required to finetune best practice, especially given the increasing use of re-irradiation.
Collapse
Affiliation(s)
- C Aristei
- Radiation Oncology Section, Department of Medicine and Surgery, University of Perugia and Perugia General Hospital, Perugia, Italy.
| | - O Kaidar-Person
- Breast Radiation Unit, Radiation Oncology, Sheba Medical Center, Ramat Gan, Israel
| | - L Boersma
- Radiation Oncology (Maastro), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - M C Leonardi
- Division of Radiation Oncology, IEO European Institute of Oncology, IRCCS, Milan, Italy
| | - B Offersen
- Department of Experimental Clinical Oncology, Department of Oncology, Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
| | - P Franco
- Depatment of Translational Medicine, University of Eastern Piedmont and Department of Radiation Oncology, 'Maggiore della Carita`' University Hospital, Novara, Italy
| | - M Arenas
- Universitat Rovira I Virgili, Radiation Oncology Department, Hospital Universitari Sant Hoan de Reus, IISPV, Spain
| | - C Bourgier
- Radiation Oncology, ICM-Val d' Aurelle, Univ Montpellier, Montpellier, France
| | - R Pfeffer
- Oncology Institute, Assuta Medical Center, Tel Aviv and Ben Gurion University Medical School, Israel
| | - V Kouloulias
- 2nd Department of Radiology, Radiotherapy Unit, Medical School, National and Kapodistrian University of Athens, Greece
| | - Y Bölükbaşı
- Koc University, Faculty of Medicine, Department of Radiation Oncology, Istanbul, Turkey
| | - I Meattini
- Department of Experimental and Clinical Biomedical Sciences "M. Serio", University of Florence & Radiation Oncology Unit - Oncology Department, Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | - C Coles
- Department of Oncology, University of Cambridge, UK
| | - A Montero Luis
- Department of Radiation Oncology, University Hospital HM Sanchinarro, HM Hospitales, Madrid, Spain
| | - V Masiello
- Unità Operativa di Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Gemelli IRCSS Roma, Italy
| | - I Palumbo
- Radiation Oncology Section, Department of Medicine and Surgery, University of Perugia and Perugia General Hospital, Perugia, Italy
| | - A G Morganti
- DIMES, Alma Mater Studiorum Bologna University, Bologna, Italy; Radiation Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum Bologna University, Bologna, Italy
| | - E Perrucci
- Radiation Oncology Section, Perugia General Hospital, Perugia, Italy
| | - V Tombolini
- Radiation Oncology, Department of Radiological, Oncological and Pathological Science, University "La Sapienza", Roma, Italy
| | - M Krengli
- DISCOG, Università di Padova e Istituto Oncologico Veneto - IRCCS, Italy
| | - F Marazzi
- Unità Operativa di Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Gemelli IRCSS Roma, Italy
| | - L Trigo
- Service of Brachytherapy, Department of Image and Radioncology, Instituto Português Oncologia Porto Francisco Gentil E.P.E., Portugal
| | - S Borghesi
- Radiation Oncology Unit of Arezzo-Valdarno, Azienda USL Toscana Sud Est, Italy
| | - A Ciabattoni
- Department of Radiation Oncology, San Filippo Neri Hospital, ASL Rome 1, Rome, Italy
| | - I Ratoša
- Division of Radiation Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia; Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - V Valentini
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Università Cattolica del Sacro Cuore e Fondazione Policlinico Gemelli IRCSS Roma, Italy
| | - P Poortmans
- University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium; Department of Radiation Oncology, Iridium Kankernetwerk, Antwerp, Belgium, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| |
Collapse
|
186
|
Hwang KP, Elshafeey NA, Kotrotsou A, Chen H, Son JB, Boge M, Mohamed RM, Abdelhafez AH, Adrada BE, Panthi B, Sun J, Musall BC, Zhang S, Candelaria RP, White JB, Ravenberg EE, Tripathy D, Yam C, Litton JK, Huo L, Thompson AM, Wei P, Yang WT, Pagel MD, Ma J, Rauch GM. A Radiomics Model Based on Synthetic MRI Acquisition for Predicting Neoadjuvant Systemic Treatment Response in Triple-Negative Breast Cancer. Radiol Imaging Cancer 2023; 5:e230009. [PMID: 37505106 PMCID: PMC10413296 DOI: 10.1148/rycan.230009] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/18/2023] [Accepted: 06/03/2023] [Indexed: 07/29/2023]
Abstract
Purpose To determine if a radiomics model based on quantitative maps acquired with synthetic MRI (SyMRI) is useful for predicting neoadjuvant systemic therapy (NAST) response in triple-negative breast cancer (TNBC). Materials and Methods In this prospective study, 181 women diagnosed with stage I-III TNBC were scanned with a SyMRI sequence at baseline and at midtreatment (after four cycles of NAST), producing T1, T2, and proton density (PD) maps. Histopathologic analysis at surgery was used to determine pathologic complete response (pCR) or non-pCR status. From three-dimensional tumor contours drawn on the three maps, 310 histogram and textural features were extracted, resulting in 930 features per scan. Radiomic features were compared between pCR and non-pCR groups by using Wilcoxon rank sum test. To build a multivariable predictive model, logistic regression with elastic net regularization and cross-validation was performed for texture feature selection using 119 participants (median age, 52 years [range, 26-77 years]). An independent testing cohort of 62 participants (median age, 48 years [range, 23-74 years]) was used to evaluate and compare the models by area under the receiver operating characteristic curve (AUC). Results Univariable analysis identified 15 T1, 10 T2, and 12 PD radiomic features at midtreatment that predicted pCR with an AUC greater than 0.70 in both the training and testing cohorts. Multivariable radiomics models of maps acquired at midtreatment demonstrated superior performance over those acquired at baseline, achieving AUCs as high as 0.78 and 0.72 in the training and testing cohorts, respectively. Conclusion SyMRI-based radiomic features acquired at midtreatment are potentially useful for identifying early NAST responders in TNBC. Keywords: MR Imaging, Breast, Outcomes Analysis ClinicalTrials.gov registration no. NCT02276443 Supplemental material is available for this article. © RSNA, 2023 See also the commentary by Houser and Rapelyea in this issue.
Collapse
Affiliation(s)
- Ken-Pin Hwang
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Nabil A. Elshafeey
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Aikaterini Kotrotsou
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Huiqin Chen
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Jong Bum Son
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Medine Boge
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Rania M. Mohamed
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Abeer H. Abdelhafez
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Beatriz E. Adrada
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Bikash Panthi
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Jia Sun
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Benjamin C. Musall
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Shu Zhang
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Rosalind P. Candelaria
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Jason B. White
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Elizabeth E. Ravenberg
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Debu Tripathy
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Clinton Yam
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Jennifer K. Litton
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Lei Huo
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Alastair M. Thompson
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Peng Wei
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Wei T. Yang
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Mark D. Pagel
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Jingfei Ma
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| | - Gaiane M. Rauch
- From the Departments of Imaging Physics (K.P.H., A.K., J.B.S., B.P.,
B.C.M., J.M.), Breast Imaging (N.A.E., M.B., R.M.M., A.H.A., B.E.A., R.P.C.,
W.T.Y., G.M.R.), Biostatistics (H.C., J.S., P.W.), Cancer Systems Imaging (S.Z.,
M.D.P.), Moon Shots Operations (J.B.W.), Breast Medical Oncology (E.E.R., D.T.,
C.Y.), Clinical Research (J.K.L.), Pathology (L.H.), and Abdominal Imaging
(G.M.R.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,
Houston, TX 77030; and Division of Surgical Oncology, Baylor College of
Medicine, Houston, Tex (A.M.T.)
| |
Collapse
|
187
|
Mohamed A, Kruse M, Tran J. Progress in immune checkpoint inhibition in early-stage triple-negative breast cancer. Expert Rev Anticancer Ther 2023; 23:1071-1084. [PMID: 37747062 DOI: 10.1080/14737140.2023.2262764] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/20/2023] [Indexed: 09/26/2023]
Abstract
INTRODUCTION Immune checkpoint inhibitors have been particularly effective in treating cancers with robust immune microenvironments and have been successfully incorporated into the management of metastatic ER-negative and HER2-negative breast cancer. This has prompted investigation of immunotherapy in early-stage triple negative breast cancer (TNBC) to address the suboptimal clinical outcomes and limited therapeutic options. AREAS COVERED This review highlights the studies examining the use of neoadjuvant immunotherapy with standard chemotherapy in the management of early-stage TNBC and explores ongoing areas of study including the role of adjuvant checkpoint inhibition and novel combination therapies with immunotherapy. EXPERT OPINION The current standard of care for early-stage ER-negative, HER2-negative breast cancer measuring ≥2 cm or with lymph node involvement is neoadjuvant chemotherapy with pembrolizumab followed by ongoing pembrolizumab in the adjuvant setting to complete 1 year of total therapy as per the KEYNOTE-522 study. This approach is associated with improved pathologic complete response (pCR) rate and event free survival, irrespective of PD-L1 status. Many questions remain regarding the optimization of chemotherapy partner(s) for immunotherapy, necessity of adjuvant immunotherapy for patients who achieve pCR, inclusion of other therapies in the adjuvant setting (particularly capecitabine or olaparib), and use of adjuvant immunotherapy when it was not received in the neoadjuvant setting.
Collapse
Affiliation(s)
- Ahmed Mohamed
- Department of Hematology and Medical Oncology, Cleveland Clinic Foundation, Taussig Cancer Institute, Cleveland, United States of America
| | - Megan Kruse
- Department of Hematology and Medical Oncology, Cleveland Clinic Foundation, Taussig Cancer Institute, Cleveland, United States of America
| | - Jennifer Tran
- Department of Hematology and Medical Oncology, Cleveland Clinic Foundation, Taussig Cancer Institute, Cleveland, United States of America
| |
Collapse
|
188
|
Guven DC, Yildirim HC, Kus F, Erul E, Kertmen N, Dizdar O, Aksoy S. Optimal adjuvant treatment strategies for TNBC patients with residual disease after neoadjuvant treatment. Expert Rev Anticancer Ther 2023; 23:1049-1059. [PMID: 37224429 DOI: 10.1080/14737140.2023.2218090] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 05/22/2023] [Indexed: 05/26/2023]
Abstract
INTRODUCTION The therapeutic armamentarium for the neoadjuvant treatment of triple-negative breast cancer (TNBC) has significantly expanded with the hopes of improving pathological complete response (pCR) rates and the possibility of a cure. However, the data on optimal adjuvant treatment strategies for patients with residual disease after neoadjuvant treatment is limited. AREAS COVERED We discuss the available data on adjuvant treatment for residual TNBC after neoadjuvant treatment considering clinical trials. Additionally, we discuss ongoing trials to give perspectives on how the field may evolve in the next decade. EXPERT OPINION The available data support the use of adjuvant capecitabine for all patients and either adjuvant capecitabine or olaparib for patients with germline BRCA1 and BRCA2 mutations, according to availability. The CREATE-X study of capecitabine and OlympiA study of olaparib demonstrated disease-free and overall survival benefits. There is an unmet need for studies comparing these two options for patients with germline BRCA mutations. Further research is needed to delineate the use of immunotherapy in the adjuvant setting, molecular targeted therapy for patients with molecular alterations other than germline BRCA mutation, combinations, and antibody-drug conjugates to further improve outcomes.
Collapse
Affiliation(s)
- Deniz Can Guven
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Hasan Cagri Yildirim
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Fatih Kus
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Enes Erul
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Neyran Kertmen
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Omer Dizdar
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Sercan Aksoy
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| |
Collapse
|
189
|
Tarekegn K, Keskinkilic M, Kristoff TJ, Evans ST, Kalinsky K. The role of immune checkpoint inhibition in triple negative breast cancer. Expert Rev Anticancer Ther 2023; 23:1095-1106. [PMID: 37771270 DOI: 10.1080/14737140.2023.2265059] [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: 07/21/2023] [Accepted: 09/26/2023] [Indexed: 09/30/2023]
Abstract
INTRODUCTION Immunotherapy has revolutionized cancer treatment, including TNBC, which has limited options of treatment and poor prognosis. ICIs studied in TNBC include pembrolizumab, nivolumab, atezolizumab, and durvalumab. Initial studies exploring ICI monotherapy demonstrated promising yet limited responses. Subsequent studies, KEYNOTE 522 and KEYNOTE 355, which combined ICI with chemotherapy, have resulted in the FDA approval of pembrolizumab in the early-stage and metastatic setting, respectively. AREAS COVERED This article provides a comprehensive review of the role of ICI in the treatment of TNBC. We reviewed the trials that have evaluated ICI monotherapy, dual therapy, ICI in combination with chemotherapy, targeted therapy, vaccines and radiation. Additionally, we reviewed potential biomarkers of response and immune-related adverse events (irAEs). A literature search was conducted via PubMed and ClinicalTrials.gov as of 5 June 2023. EXPERT OPINION Various approaches combining immunotherapy with chemotherapy, targeted therapy, vaccines and radiation have been assessed. Pembrolizumab remains the only ICI approved in both the early stage and mTNBC. The role of adjuvant pembrolizumab in those who achieved pCR after neoadjuvant therapy is being investigated. Combining ICI with PARP inhibitors and radiation shows promise. More research is needed in identifying predictors of response. Monitoring of irAEs remains crucial.
Collapse
Affiliation(s)
- Kidist Tarekegn
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Merve Keskinkilic
- Department of Medical Oncology, Dokuz Eylül University Faculty of Medicine, Izmir, Turkey
| | | | - Sean T Evans
- Emory University School of Medicine, Atlanta, GA, USA
| | - Kevin Kalinsky
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| |
Collapse
|
190
|
Kotteas E, Bielo LB, Valenza C, Santoro C, Koukoutzeli C, Trapani D, Curigliano G. Treatment optimization in early triple negative breast cancer. Expert Rev Anticancer Ther 2023; 23:1107-1116. [PMID: 37873652 DOI: 10.1080/14737140.2023.2268840] [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: 08/10/2023] [Accepted: 10/05/2023] [Indexed: 10/25/2023]
Abstract
INTRODUCTION The treatment of early-stage triple-negative breast cancer (TNBC) has radically changed in recent years. Response to neoadjuvant treatment has provided prognostic information, and the achievement of a pathological complete response (pCR) is associated with improved prognosis. An exact treatment algorithm that embraces the trade-off of efficacy and toxicity in a risk-adapted manner has, however, not been consolidated. AREAS COVERED In this review, we focused on the current treatments used for patients with early triple negative breast cancer, aiming at framing a therapeutic approach toward risk-adapted treatment optimization. We reviewed the clinical trials and other evidence at the foundation of the current clinical practice in early TNBC and identified possible areas of clinical implementation. EXPERT OPINION In our opinion, treatment optimization will ensure improved patient-centric outcomes, with less toxicities, better long-term quality of life and risk-adapted treatment modulation. Presently, treatment modulation can be applied in some patients through de-intensification, for small TNBC, informed by novel biomarkers and based on the response to neoadjuvant treatments, especially in the case of pCR. Innovative approaches should incorporate baseline risk and cancer biology, treatment response, and post-surgery biomarkers of prognosis, to deliver risk-adapted treatments for patients with early TNBC.
Collapse
Affiliation(s)
- Elias Kotteas
- Oncology Unit, 3rd Department of Internal Medicine, National & Kapodistrian University of Athens, Athens, Greece
| | - Luca Boscolo Bielo
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Carmine Valenza
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Celeste Santoro
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Chrysanthi Koukoutzeli
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
| | - Dario Trapani
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
191
|
Johnson HM, Valero V, Yang WT, Smith BD, Krishnamurthy S, Shen Y, Lin H, Lucci A, Rauch GM, Kuerer HM. Eliminating Breast Surgery for Invasive Cancer with Exceptional Response to Neoadjuvant Systemic Therapy: Prospective Multicenter Clinical Trial Planned Initial Feasibility Endpoint. J Am Coll Surg 2023; 237:101-108. [PMID: 36856291 PMCID: PMC11892649 DOI: 10.1097/xcs.0000000000000670] [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] [Indexed: 03/02/2023]
Abstract
BACKGROUND Response to neoadjuvant systemic therapy (NST) for breast cancer enables tailoring of subsequent therapy. Image-guided breast biopsy after NST can accurately predict a pathologic complete response (pCR). The feasibility phase of the clinical trial reported here assesses omission of breast surgery followed by radiotherapy in terms of local recurrence before trial expansion. STUDY DESIGN Women with unicentric, cT1-2 N0-1 M0 triple-negative (TNBC) or human epidermal growth factor receptor 2-positive breast cancer (HER2+BC) cancer with <2 cm residual disease on post-NST imaging were eligible to enroll. If no residual invasive or in situ disease was identified by image-guided, vacuum-assisted core biopsy (VACB), breast surgery was omitted, and radiotherapy delivered. The primary endpoint for the feasibility phase was ipsilateral breast tumor recurrence at 6 months. If any recurrence occurred during the feasibility phase the trial would halt. RESULTS Thirteen patients were enrolled from March 2017 to October 2018. The mean age was 60.8 years (range 51 to 75) and most patients were White (69.2%) and non-Hispanic/Latino (84.6%). All patients had invasive ductal carcinoma (6 TNBC, 7 HER2+BC). Mean tumor size was 2.4 cm (range 0.9 to 5.0) before NST and 0.7 cm (range 0 to 1.8) after NST. Seven patients (53.8%) had residual disease identified on VACB; the remaining 6 (46.2%) comprised the feasibility cohort. At a median follow-up of 44.3 months (range 41.3 to 51.3) there was no ipsilateral breast tumor recurrence in this cohort. CONCLUSIONS These early data suggest that omission of breast surgery in patients with invasive TNBC and HER2+BC with no evidence of residual disease on standardized VACB after NST is potentially feasible. Results from the expansion phase of this clinical trial will be reported per protocol prespecified analyses.
Collapse
Affiliation(s)
- Helen M Johnson
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vicente Valero
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wei T Yang
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Benjamin D Smith
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Savitri Krishnamurthy
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yu Shen
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Heather Lin
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anthony Lucci
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gaiane M Rauch
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Abdominal Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Henry M Kuerer
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
192
|
He ZY, Zhuo RG, Yang SP, Zhou P, Xu JY, Zhou J, Wu SG. CircNCOR1 regulates breast cancer radiotherapy efficacy by regulating CDK2 via hsa-miR-638 binding. Cell Signal 2023:110787. [PMID: 37391048 DOI: 10.1016/j.cellsig.2023.110787] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/19/2023] [Accepted: 06/27/2023] [Indexed: 07/02/2023]
Abstract
BACKGROUND Despite aggressive local and regional therapy, triple-negative breast cancer (TNBC) is characterized by an increased risk of locoregional recurrence. RNA-sequencing data has identified a large number of circRNAs in primary breast cancers, but the role of specific circRNAs in regulating the radiosensitivity of TNBC is not fully understood. This research aimed to investigate the function of circNCOR1 in the radiosensitivity of TNBC. METHODS CircRNA high-throughput sequencing was conducted on two breast cancer MDA-MB-231 and BT549 cell lines after 6 Gy radiation. The relationship between circNCOR1, hsa-miR-638, and CDK2 was determined by RNA immunoprecipitation (RIP), FISH and luciferase assays. The proliferation and apoptosis of breast cancer cells were measured by CCK8, flow cytometry, colony formation assays, and western blot. RESULTS Differential expression of circRNAs was closely related to the proliferation of breast cancer cells after irradiation. Overexpression of circNCOR1 facilitated the proliferation of MDA-MB-231 and BT549 cells and impaired the radiosensitivity of breast cancer cells. Additionally, circNCOR1 acted as a sponge for hsa-miR-638 to regulate the downstream target protein CDK2. Overexpression of hsa-miR-638 promoted apoptosis of breast cancer cells, while overexpression of CDK2 alleviated apoptosis and increased proliferation and clonogenicity. In vivo, overexpression of circNCOR1 partially reversed radiation-induced loosening of tumor structures and enhanced tumor cell proliferation. CONCLUSION Our results demonstrated that circNCOR1 bounds to hsa-miR-638 and targets CDK2, thereby regulating the radiosensitivity of TNBC.
Collapse
Affiliation(s)
- Zhen-Yu He
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, People's Republic of China
| | - Ren-Gong Zhuo
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen 361102, People's Republic of China
| | - Shi-Ping Yang
- Department of Radiation Oncology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou 570311, People's Republic of China
| | - Ping Zhou
- Department of Radiation Oncology, Xiamen Cancer Center, Xiamen Key Laboratory of Radiation Oncology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, People's Republic of China
| | - Jing-Ying Xu
- Department of Obstetrics and Gynecology, Xiamen Cancer Center, Xiamen Key Laboratory of Radiation Oncology, The First Affiliated Hospital of Xiamen University, Xiamen 361003, People's Republic of China
| | - Juan Zhou
- Department of Obstetrics and Gynecology, Xiamen Cancer Center, Xiamen Key Laboratory of Radiation Oncology, The First Affiliated Hospital of Xiamen University, Xiamen 361003, People's Republic of China.
| | - San-Gang Wu
- Department of Radiation Oncology, Xiamen Cancer Center, Xiamen Key Laboratory of Radiation Oncology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, People's Republic of China.
| |
Collapse
|
193
|
Page DB, Pucilowska J, Chun B, Kim I, Sanchez K, Moxon N, Mellinger S, Wu Y, Koguchi Y, Conrad V, Redmond WL, Martel M, Sun Z, Campbell MB, Conlin A, Acheson A, Basho R, McAndrew P, El-Masry M, Park D, Bennetts L, Seitz RS, Nielsen TJ, McGregor K, Rajamanickam V, Bernard B, Urba WJ, McArthur HL. A phase Ib trial of pembrolizumab plus paclitaxel or flat-dose capecitabine in 1st/2nd line metastatic triple-negative breast cancer. NPJ Breast Cancer 2023; 9:53. [PMID: 37344474 DOI: 10.1038/s41523-023-00541-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 04/21/2023] [Indexed: 06/23/2023] Open
Abstract
Chemoimmunotherapy with anti-programmed cell death 1/ligand 1 and cytotoxic chemotherapy is a promising therapeutic modality for women with triple-negative breast cancer, but questions remain regarding optimal chemotherapy backbone and biomarkers for patient selection. We report final outcomes from a phase Ib trial evaluating pembrolizumab (200 mg IV every 3 weeks) with either weekly paclitaxel (80 mg/m2 weekly) or flat-dose capecitabine (2000 mg orally twice daily for 7 days of every 14-day cycle) in the 1st/2nd line setting. The primary endpoint is safety (receipt of 2 cycles without grade III/IV toxicities requiring discontinuation or ≥21-day delays). The secondary endpoint is efficacy (week 12 objective response). Exploratory aims are to characterize immunologic effects of treatment over time, and to evaluate novel biomarkers. The trial demonstrates that both regimens meet the pre-specified safety endpoint (paclitaxel: 87%; capecitabine: 100%). Objective response rate is 29% for pembrolizumab/paclitaxel (n = 4/13, 95% CI: 10-61%) and 43% for pembrolizumab/capecitabine (n = 6/14, 95% CI: 18-71%). Partial responses are observed in two subjects with chemo-refractory metaplastic carcinoma (both in capecitabine arm). Both regimens are associated with significant peripheral leukocyte contraction over time. Response is associated with clinical PD-L1 score, non-receipt of prior chemotherapy, and the H&E stromal tumor-infiltrating lymphocyte score, but also by a novel 27 gene IO score and spatial biomarkers (lymphocyte spatial skewness). In conclusion, pembrolizumab with paclitaxel or capecitabine is safe and clinically active. Both regimens are lymphodepleting, highlighting the competing immunostimulatory versus lymphotoxic effects of cytotoxic chemotherapy. Further exploration of the IO score and spatial TIL biomarkers is warranted. The clinical trial registration is NCT02734290.
Collapse
Affiliation(s)
- David B Page
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA.
| | - Joanna Pucilowska
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Brie Chun
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Isaac Kim
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Katherine Sanchez
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Nicole Moxon
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Staci Mellinger
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Yaping Wu
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Yoshinobu Koguchi
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Valerie Conrad
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - William L Redmond
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Maritza Martel
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Zhaoyu Sun
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Mary B Campbell
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Alison Conlin
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Anupama Acheson
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Reva Basho
- Cedars Sinai Medical Center, Los Angeles, CA, USA
- Ellison Institute for Transformative Medicine, Los Angeles, CA, USA
| | | | | | - Dorothy Park
- Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Laura Bennetts
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | | | | | | | | | - Brady Bernard
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Walter J Urba
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Heather L McArthur
- Cedars Sinai Medical Center, Los Angeles, CA, USA
- UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
194
|
Yu K, Basu A, Yau C, Wolf DM, Goodarzi H, Bandyopadhyay S, Korkola JE, Hirst GL, Asare S, DeMichele A, Hylton N, Yee D, Esserman L, van ‘t Veer L, Sirota M. Computational drug repositioning for the identification of new agents to sensitize drug-resistant breast tumors across treatments and receptor subtypes. Front Oncol 2023; 13:1192208. [PMID: 37384294 PMCID: PMC10294228 DOI: 10.3389/fonc.2023.1192208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/25/2023] [Indexed: 06/30/2023] Open
Abstract
Introduction Drug resistance is a major obstacle in cancer treatment and can involve a variety of different factors. Identifying effective therapies for drug resistant tumors is integral for improving patient outcomes. Methods In this study, we applied a computational drug repositioning approach to identify potential agents to sensitize primary drug resistant breast cancers. We extracted drug resistance profiles from the I-SPY 2 TRIAL, a neoadjuvant trial for early stage breast cancer, by comparing gene expression profiles of responder and non-responder patients stratified into treatments within HR/HER2 receptor subtypes, yielding 17 treatment-subtype pairs. We then used a rank-based pattern-matching strategy to identify compounds in the Connectivity Map, a database of cell line derived drug perturbation profiles, that can reverse these signatures in a breast cancer cell line. We hypothesize that reversing these drug resistance signatures will sensitize tumors to treatment and prolong survival. Results We found that few individual genes are shared among the drug resistance profiles of different agents. At the pathway level, however, we found enrichment of immune pathways in the responders in 8 treatments within the HR+HER2+, HR+HER2-, and HR-HER2- receptor subtypes. We also found enrichment of estrogen response pathways in the non-responders in 10 treatments primarily within the hormone receptor positive subtypes. Although most of our drug predictions are unique to treatment arms and receptor subtypes, our drug repositioning pipeline identified the estrogen receptor antagonist fulvestrant as a compound that can potentially reverse resistance across 13/17 of the treatments and receptor subtypes including HR+ and triple negative. While fulvestrant showed limited efficacy when tested in a panel of 5 paclitaxel resistant breast cancer cell lines, it did increase drug response in combination with paclitaxel in HCC-1937, a triple negative breast cancer cell line. Conclusion We applied a computational drug repurposing approach to identify potential agents to sensitize drug resistant breast cancers in the I-SPY 2 TRIAL. We identified fulvestrant as a potential drug hit and showed that it increased response in a paclitaxel-resistant triple negative breast cancer cell line, HCC-1937, when treated in combination with paclitaxel.
Collapse
Affiliation(s)
- Katharine Yu
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Amrita Basu
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Christina Yau
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Denise M. Wolf
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Hani Goodarzi
- University of California, San Francisco, San Francisco, CA, United States
| | | | - James E. Korkola
- Oregon Health and Science University, Portland, OR, United States
| | - Gillian L. Hirst
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Smita Asare
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States
- QuantumLeap Healthcare Collaborative, San Francisco, CA, United States
| | | | - Nola Hylton
- University of California, San Francisco, San Francisco, CA, United States
| | - Douglas Yee
- University of Minnesota, Minneapolis, MN, United States
| | - Laura Esserman
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Laura van ‘t Veer
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
195
|
Zaborowski AM, Wong SM. Neoadjuvant systemic therapy for breast cancer. Br J Surg 2023; 110:765-772. [PMID: 37104057 PMCID: PMC10683941 DOI: 10.1093/bjs/znad103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 04/02/2023] [Indexed: 04/28/2023]
Affiliation(s)
| | - Stephanie M Wong
- Department of Surgery and Oncology, McGill University Medical School, Montreal, Quebec, Canada
- Segal Cancer Centre, Sir Mortimer B. Davis Jewish General Hospital, Montreal, Quebec, Canada
| |
Collapse
|
196
|
Downs-Canner S, Mittendorf EA. Preoperative Immunotherapy Combined with Chemotherapy for Triple-Negative Breast Cancer: Perspective on the KEYNOTE-522 Study. Ann Surg Oncol 2023; 30:3166-3169. [PMID: 36897418 DOI: 10.1245/s10434-023-13267-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 02/09/2023] [Indexed: 03/11/2023]
Affiliation(s)
- Stephanie Downs-Canner
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elizabeth A Mittendorf
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA.
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
197
|
Parker BA, Shatsky RA, Schwab RB, Wallace AM, Wolf DM, Hirst GL, Brown-Swigart L, Esserman LJ, van 't Veer LJ, Ghia EM, Yau C, Kipps TJ. Association of baseline ROR1 and ROR2 gene expression with clinical outcomes in the I-SPY2 neoadjuvant breast cancer trial. Breast Cancer Res Treat 2023; 199:281-291. [PMID: 37029329 PMCID: PMC10175386 DOI: 10.1007/s10549-023-06914-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 03/12/2023] [Indexed: 04/09/2023]
Abstract
PURPOSE ROR1 and ROR2 are Type 1 tyrosine kinase-like orphan receptors for Wnt5a that are associated with breast cancer progression. Experimental agents targeting ROR1 and ROR2 are in clinical trials. This study evaluated whether expression levels of ROR1 or ROR2 correlated with one another or with clinical outcomes. METHODS We interrogated the clinical significance of high-level gene expression of ROR1 and/or ROR2 in the annotated transcriptome dataset from 989 patients with high-risk early breast cancer enrolled in one of nine completed/graduated/experimental and control arms in the neoadjuvant I-SPY2 clinical trial (NCT01042379). RESULTS High ROR1 or high ROR2 was associated with breast cancer subtypes. High ROR1 was more prevalent among hormone receptor-negative and human epidermal growth factor receptor 2-negative (HR-HER2-) tumors and high ROR2 was less prevalent in this subtype. Although not associated with pathologic complete response, high ROR1 or high ROR2 each was associated with event-free survival (EFS) in distinct subtypes. High ROR1 associated with a worse EFS in HR + HER2- patients with high post-treatment residual cancer burden (RCB-II/III) (HR 1.41, 95% CI = 1.11-1.80) but not in patients with minimal post-treatment disease (RCB-0/I) (HR 1.85, 95% CI = 0.74-4.61). High ROR2 associated with an increased risk of relapse in patients with HER2 + disease and RCB-0/I (HR 3.46, 95% CI = 1.33-9.020) but not RCB-II/III (HR 1.07, 95% CI = 0.69-1.64). CONCLUSION High ROR1 or high ROR2 distinctly identified subsets of breast cancer patients with adverse outcomes. Further studies are warranted to determine if high ROR1 or high ROR2 may identify high-risk populations for studies of targeted therapies.
Collapse
Affiliation(s)
- Barbara A Parker
- Department of Medicine and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA.
| | - Rebecca A Shatsky
- Department of Medicine and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Richard B Schwab
- Department of Medicine and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Anne M Wallace
- Department of Surgery and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Denise M Wolf
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Gillian L Hirst
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Lamorna Brown-Swigart
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Laura J Esserman
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Laura J van 't Veer
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Emanuela M Ghia
- Department of Medicine and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
- Center for Novel Therapeutics, University of California San Diego, La Jolla, CA, USA
| | - Christina Yau
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Thomas J Kipps
- Department of Medicine and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
- Center for Novel Therapeutics, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
198
|
Ali M, Wood S, Pryor D, Moon D, Bressel M, Azad AA, Mitchell C, Murphy D, Zargar H, Hardcastle N, Kearsley J, Eapen R, Wong LM, Cuff K, Lawrentschuk N, Neeson PJ, Siva S. NeoAdjuvant pembrolizumab and STEreotactic radiotherapy prior to nephrectomy for renal cell carcinoma (NAPSTER): A phase II randomised clinical trial. Contemp Clin Trials Commun 2023; 33:101145. [PMID: 37168818 PMCID: PMC10164766 DOI: 10.1016/j.conctc.2023.101145] [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: 08/16/2022] [Revised: 04/13/2023] [Accepted: 04/21/2023] [Indexed: 05/13/2023] Open
Abstract
Background Surgery remains the standard of care for localised renal cell carcinoma (RCC). Nevertheless, nearly 50% of patients with high-risk disease experience relapse after surgery, with distant sites being common. Considering improved outcomes in terms of disease-free survival with adjuvant immunotherapy with pembrolizumab, we hypothesise that neoadjuvant SABR with or without the addition of pembrolizumab before nephrectomy will lead to improved disease outcomes by evoking better immune response in the presence of an extensive reserve of tumor-associated antigens. Methods and analysis This prospective, open-label, phase II, randomised, non-comparative, clinical trial will investigate the use of neoadjuvant stereotactic ablative body radiotherapy (SABR) with or without pembrolizumab prior to nephrectomy. The trial will be conducted at two centres in Australia that are well established for delivering SABR to primary RCC patients. Twenty-six patients with biopsy-proven clear cell RCC will be recruited over two years. Patients will be randomised to either SABR or SABR/pembrolizumab. Patients in both arms will undergo surgery at 9 weeks after completion of experimental treatment. The primary objectives are to describe major pathological response and changes in tumour-responsive T-cells from baseline pre-treatment biopsy in each arm. Patients will be followed for sixty days post-surgery. Outcomes and significance We hypothesize that SABR alone or SABR plus pembrolizumab will induce significant tumor-specific immune response and major pathological response. In that case, either one or both arms could justifiably be used as a neoadjuvant treatment approach in future randomized trials in the high-risk patient population.
Collapse
Affiliation(s)
- Muhammad Ali
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Simon Wood
- Metro South Hospital and Health Service, Brisbane, QLD, Australia
- Department of Urology, Princess Alexandra Hospital, Brisbane, QLD, Australia
- Centre for Kidney Disease Research, Translational Research Institute, Brisbane, QLD, Australia
| | - David Pryor
- Department of Radiation Oncology, Princess Alexandra Hospital, Brisbane, QLD, Australia
- Queensland University of Technology, Brisbane, QLD, Australia
| | - Daniel Moon
- Deapartment of Surgery, The University of Melbourne, Melbourne, VIC, Australia
| | - Mathias Bressel
- Centre for Biostatistics and Clinical Trials, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Arun A. Azad
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Catherine Mitchell
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Declan Murphy
- Department of Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Homi Zargar
- Deapartment of Surgery, The University of Melbourne, Melbourne, VIC, Australia
| | - Nick Hardcastle
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
- Department of Physical Sciences, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Centre for Medical Radiation Physics, University of Wollongong, NSW, Australia
| | - Jamie Kearsley
- Department of Urology, Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Renu Eapen
- Department of Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Lih Ming Wong
- Deapartment of Surgery, The University of Melbourne, Melbourne, VIC, Australia
- Department of Urology, St Vincent's Health, Melbourne, VIC, Australia
| | - Katharine Cuff
- Department of Medical Oncology, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Nathan Lawrentschuk
- Deapartment of Surgery, The University of Melbourne, Melbourne, VIC, Australia
- Department of Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Department of Urology, Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Paul J. Neeson
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Shankar Siva
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
199
|
Han HS, Vikas P, Costa RLB, Jahan N, Taye A, Stringer-Reasor EM. Early-Stage Triple-Negative Breast Cancer Journey: Beginning, End, and Everything in Between. Am Soc Clin Oncol Educ Book 2023; 43:e390464. [PMID: 37335956 DOI: 10.1200/edbk_390464] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2023]
Abstract
Triple-negative breast cancer (TNBC) is a very heterogeneous and aggressive breast cancer subtype with a high risk of mortality, even if diagnosed early. The mainstay of early-stage breast cancer includes systemic chemotherapy and surgery, with or without radiation therapy. More recently, immunotherapy is approved to treat TNBC, but managing immune-rated adverse events while balancing efficacy is a challenge. The purpose of this review is to highlight the current treatment recommendations for early-stage TNBC and the management of immunotherapy toxicities.
Collapse
Affiliation(s)
- Hyo Sook Han
- Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Praveen Vikas
- The University of Iowa Holden Comprehensive Cancer Center, Iowa City, IA
| | - Ricardo L B Costa
- Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Nusrat Jahan
- Department of Medicine, Division of Hematology Oncology, University of Alabama at Birmingham, O'Neal Comprehensive Cancer Center, Birmingham, AL
| | - Ammanuel Taye
- Department of Medicine, Division of Hematology Oncology, University of Alabama at Birmingham, O'Neal Comprehensive Cancer Center, Birmingham, AL
| | - Erica M Stringer-Reasor
- Department of Medicine, Division of Hematology Oncology, University of Alabama at Birmingham, O'Neal Comprehensive Cancer Center, Birmingham, AL
| |
Collapse
|
200
|
Chen J, Han Y, Hu Y, Feng X, Meng X, Guo S, Sun C, Chen G, Li K. Neoadjuvant camrelizumab plus chemotherapy for locally advanced cervical cancer (NACI Study): a study protocol of a prospective, single-arm, phase II trial. BMJ Open 2023; 13:e067767. [PMID: 37253491 PMCID: PMC10254908 DOI: 10.1136/bmjopen-2022-067767] [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: 09/06/2022] [Accepted: 05/14/2023] [Indexed: 06/01/2023] Open
Abstract
INTRODUCTION Neoadjuvant chemotherapy (NACT) is an emerging approach for locally advanced cervical cancer (LACC). However, the clinical response and postoperative adjuvant radiation or chemoradiation trimodality treatment resulted in controversy. PD-1 inhibitors have shown promising role in recurrent or metastatic cervical cancer, and there is preclinical evidence of the activation and synergistic effects of NACT on PD-1 inhibitors. This study aims to evaluate the efficacy and safety of the preoperative PD-1 inhibitor camrelizumab combined with NACT for LACC. METHODS AND ANALYSIS The study is designed as a multicentre, open-label, single-arm, prospective phase II study. A total of 82 patients will receive neoadjuvant chemo-immunotherapy, defined as one cycle of cisplatin (75-80 mg/m2, intravenously) plus nab-paclitaxel (260 mg/m2, intravenously) NACT and subsequent two cycles of camrelizumab (200 mg, intravenously) combined with NACT. After neoadjuvant chemo-immunotherapy, patients exhibiting complete response and partial response will undergo radical surgery and subsequent adjuvant therapy. In contrast, patients with stable disease and progressive disease will transfer to concurrent chemoradiotherapy (CCRT). Following surgery, patients will receive adjuvant CCRT or radiotherapy. The primary endpoint is the objective response rate. The secondary endpoints are the pathological complete response, patients requiring postoperative adjuvant therapy, safety of neoadjuvant chemo-immunotherapy, surgical complication, event-free survival, and overall survival. An additional aim is to dynamically evaluate peripheral immune responses and local immunological microenvironments and their association with neoadjuvant immunotherapy. ETHICS AND DISSEMINATION This trial was approved by the Medical Ethics Committee of Tongji Medical College, Huazhong University of Science and Technology (S2020-112). This study is among the first to evaluate the efficacy and safety of neoadjuvant chemo-immunotherapy in LACC. The findings of this research will promote neoadjuvant anti-PD-1 immunotherapy with radical surgery as a new therapeutic strategy. TRIAL REGISTRATION NUMBER ClinicalTrials.gov Registry (NCT04516616).
Collapse
Affiliation(s)
- Jing Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yingyan Han
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yingjie Hu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xue Feng
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xiaolin Meng
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Shuaiqingying Guo
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Chaoyang Sun
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Gang Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Kezhen Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|