1
|
Pires MCDS, Sobreira-da-Silva MJ, de Araújo PP, Retto MPF. Use of medications in women with triple-negative breast cancer between 2018 and 2019 in a Brazilian public hospital: a retrospective study. EPIDEMIOLOGIA E SERVIÇOS DE SAÚDE 2025; 34:e20240180. [PMID: 40008715 PMCID: PMC11845119 DOI: 10.1590/s2237-96222025v34e20240180.en] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 10/25/2024] [Indexed: 02/27/2025] Open
Abstract
OBJECTIVE To describe the profile of medication use in women with triple-negative breast cancer treated between 2018 and 2019 in a Brazilian public hospital. METHODS Descriptive and retrospective study, with data obtained from the Hospital Cancer Registry and physical and electronic medical records from a public hospital that is a reference in cancer treatment, in Rio de Janeiro. Descriptive analyses and analyses of time to treatment failure and overall survival were performed using the Kaplan Meier method. RESULTS Of the 176 patients, 39.0% were under 50 years of age and 47.7% were diagnosed at an advanced stage. Use of 12 chemotherapy regimens was identified, with neoadjuvant or adjuvant intent, for treatment of triple-negative breast cancer. The most commonly used treatment regimen included doxorubicin, cyclophosphamide and taxanes (docetaxel or paclitaxel). After 180 days, 76.1% of patients remained on the initial treatment. Average time until treatment failure was 7.6 months for those who followed the main regimen. Median overall survival was 34 months, and 55.7% of patients died by the end of the follow-up period (48 months). CONCLUSION The results showed that treatment with doxorubicin, cyclophosphamide and taxanes (docetaxel or paclitaxel) was the most used in the patients analyzed, that average time to treatment failure using this regimen was less than one year and that more than half of the patients died within four years after diagnosis.
Collapse
|
2
|
Zhu B, Cai Y, Zhou L, Zhao L, Chen J, Shan X, Sun X, You Q, Gong X, Zhang W, Zhu HH, Zhang P, Li Y. Injectable supramolecular hydrogel co-loading abemaciclib/NLG919 for neoadjuvant immunotherapy of triple-negative breast cancer. Nat Commun 2025; 16:687. [PMID: 39814714 PMCID: PMC11735626 DOI: 10.1038/s41467-025-55904-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/04/2025] [Indexed: 01/18/2025] Open
Abstract
The efficacy of cancer immunotherapy relies on a sufficient amount of functional immune cells. Triple-negative breast cancer lacks enough immune cell infiltration, and adjuvant therapy is necessary to prime anti-tumor immunity. However, the improvement in efficacy is unsatisfactory with concern about inducing systemic immunotoxicity. Herein, we create an abemaciclib-loaded supramolecular peptide hydrogel formed by peptide-drug amphiphiles for neoadjuvant immunotherapy of triple-negative breast cancer, where the amphiphile is a conjugate of a β-sheet-forming peptide with 1-cyclohexyl-2-(5H-imidazo[5,1-a]isoindol-5-yl)ethanol (NLG919), an inhibitor of indoleamine 2,3-dioxygenase 1. The hydrogel can be injected into the tumor site and retained for at least one week for the sustained release of both abemaciclib and NLG919. The abemaciclib is able to induce immunogenic cell death of cancer cells and increase interleukin-2 secretion by cytotoxic T lymphocytes. Abemaciclib adversely upregulates indoleamine 2,3-dioxygenase 1, whose kynurenine production activity is inhibited by NLG919. The neoadjuvant immunotherapy reduces tumor recurrence and pulmonary metastasis and prolongs the survival of animals. This hydrogel provides a potential platform for neoadjuvant immunotherapy of triple-negative breast cancer with reduced toxicity compared with free abemaciclib.
Collapse
Affiliation(s)
- Binyu Zhu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, China
| | - Ying Cai
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Pharmaceutical Science, Shandong, China
| | - Lingli Zhou
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lei Zhao
- School of Chemical Engineering, Zhengzhou University, Zhengzhou, China
| | - Jiameng Chen
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, China
| | - Xiaoting Shan
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, China
| | - Xujie Sun
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, China
| | - Qian You
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Stem Cell Research Center, Department of Urology, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Xiang Gong
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, Shanghai, China
| | - Wen Zhang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, Shanghai, China
| | - Helen He Zhu
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Stem Cell Research Center, Department of Urology, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Pengcheng Zhang
- School of Biomedical Engineering & State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai, China.
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, China.
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Pharmaceutical Science, Shandong, China.
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.
- Shandong Laboratory of Yantai Drug Discovery, Bohai rim Advanced Research Institute for Drug Discovery, Shandong, China.
| |
Collapse
|
3
|
Dugo M, Huang CS, Egle D, Bermejo B, Zamagni C, Seitz RS, Nielsen TJ, Thill M, Antón-Torres A, Russo S, Ciruelos EM, Schweitzer BL, Ross DT, Galbardi B, Greil R, Semiglazov V, Gyorffy B, Colleoni M, Kelly CM, Mariani G, Del Mastro L, Blasi O, Callari M, Pusztai L, Valagussa P, Viale G, Gianni L, Bianchini G. The Immune-Related 27-Gene Signature DetermaIO Predicts Response to Neoadjuvant Atezolizumab plus Chemotherapy in Triple-Negative Breast Cancer. Clin Cancer Res 2024; 30:4900-4909. [PMID: 39308141 PMCID: PMC11528202 DOI: 10.1158/1078-0432.ccr-24-0149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/22/2024] [Accepted: 08/26/2024] [Indexed: 11/02/2024]
Abstract
PURPOSE We assessed the 27-gene RT-qPCR-based DetermaIO assay and the same score calculated from RNA sequencing (RNA-seq) data as predictors of sensitivity to immune checkpoint therapy in the neoTRIPaPDL1 randomized trial that compared neoadjuvant carboplatin/nab-paclitaxel chemotherapy (CT) plus atezolizumab with CT alone in stage II/III triple-negative breast cancer. We also assessed the predictive function of the immuno-oncology (IO) score in expression data of patients treated with pembrolizumab plus paclitaxel (N = 29) or CT alone (N = 56) in the I-SPY2 trial. EXPERIMENTAL DESIGN RNA-seq data were obtained from pretreatment core biopsies from 242 (93.8%) of the 258 patients in the per-protocol-population. The DetermaIO RT-qPCR test, performed in the CAP/CLIA-accredited laboratory of Oncocyte Corp., was available for 220 patients (85.3%). A previously established threshold was used to assign DetermaIO-positive versus DetermaIO-negative status. Publicly available microarray data were used from I-SPY2. RESULTS IO scores calculated from RNA-seq and RT-qPCR data were highly concordant. In neoTRIPaPDL1, DetermaIO-positive cancers (N = 92, 41.8%) had pathologic complete response (pCR) rates of 69.8% and 46.9% in the CT + atezolizumab and CT arms, respectively. In DetermaIO-negative cases, pCR rates were similar in both arms (44.6% vs. 49.2%; interaction test P = 0.04). PDL1 protein expression and stromal tumor-infiltrating lymphocyte count were not predictive of differential benefit from atezolizumab. In I-SPY2, IO-positive cancers (45.9%) had pCR rates of 85.7% and 16%, with and without immunotherapy, respectively. In IO-negative cancers, pCR rates were 46.7% versus 16.1%. CONCLUSIONS DetermaIO identified patients who benefited from neoadjuvant immunotherapy resulting in improved pCR rate, independently of PDL1.
Collapse
Affiliation(s)
- Matteo Dugo
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Chiun-Sheng Huang
- National Taiwan University Hospital, College of Medicine, National Taiwan University and Taiwan Breast Cancer Consortium, Taipei, Taiwan
| | - Daniel Egle
- Department of Gynecology, Brust Gesundheit Zentrum Tirol, Medical University Innsbruck, Innsbruck, Austria
| | - Begoña Bermejo
- Medical Oncology, Hospital Clínico Universitario de Valencia, Biomedical Research Institute INCLIVA, Valencia, Spain
| | - Claudio Zamagni
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | | | | | - Marc Thill
- Department of Gynecology and Gynecological Oncology, Agaplesion Markus Krankenhaus, Frankfurt, Germany
| | | | - Stefania Russo
- Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
| | | | | | | | - Barbara Galbardi
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Richard Greil
- Department of Internal Medicine III, Paracelsus Medical University Salzburg, Salzburg Cancer Research Institute-CCCIT; and Cancer Cluster Salzburg, Salzburg, Austria
| | - Vladimir Semiglazov
- N. N. Petrov Research Institute of Oncology, St. Petersburg, Russian Federation
| | - Balázs Gyorffy
- Department of Bioinformatics, Semmelweis University, Budapest, Hungary
- Department of Biophysics, Medical School, University of Pecs, Pecs, Hungary
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
| | | | - Catherine M Kelly
- MaterMater Private Hospital, Dublin and Breast Group Cancer Trials Ireland, Dublin, Ireland
| | | | - Lucia Del Mastro
- University of Genova; IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Olivia Blasi
- IEO, Istituto Europeo di Oncologia IRCCS, Milan, Italy
| | | | - Lajos Pusztai
- Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut
| | | | | | | | - Giampaolo Bianchini
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
4
|
Takahashi S, Sato N, Kaneko K, Masuda N, Kawai M, Hirakawa H, Nomizu T, Iwata H, Ueda A, Ishikawa T, Bando H, Inoue Y, Ueno T, Ohno S, Kubo M, Yamauchi H, Okamoto M, Tokunaga E, Kamigaki S, Aogi K, Komatsu H, Kitada M, Uemoto Y, Toyama T, Yamamoto Y, Yamashita T, Yanagawa T, Yamashita H, Matsumoto Y, Toi M, Miyashita M, Ishida T, Fujishima F, Sato S, Yamaguchi T, Takahashi F, Ishioka C. TP53 signature predicts pathological complete response after neoadjuvant chemotherapy for breast cancer: Observational and confirmational study using prospective study cohorts. Transl Oncol 2024; 48:102060. [PMID: 39047382 PMCID: PMC11325231 DOI: 10.1016/j.tranon.2024.102060] [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: 05/04/2024] [Revised: 06/17/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024] Open
Abstract
The TP53 signature is considered a predictor of neoadjuvant chemotherapy (NAC) response and prognostic factor in breast cancer. The objective of this study was to confirm TP53 signature can predict pathological complete response (pCR) and prognosis in cohorts of breast cancer patients who received NAC in prospective studies. Development cohorts (retrospective [n = 37] and prospective [n = 216] cohorts) and validation cohorts (NAC administered prospective study cohorts [n = 407] and retrospective perioperative chemotherapy (PC)-naïve, hormone receptor (HrR)-positive cohort [PC-naïve_HrR+ cohort] [n = 322]) were used. TP53 signature diagnosis kit was developed using the development cohorts. TP53 signature predictability for pCR and the relationship between recurrence-free survival (RFS), overall survival (OS), and the TP53 signature were analyzed. The pCR rate of the mutant (mt) signature group was significantly higher than that of the wild-type (wt) signature group (odds ratio, 5.599; 95 % confidence interval = 1.876-16.705; P = 0.0008). The comparison of the RFS and OS between the HrR+ and HER2- subgroup of the NAC cohort and of the PC-naïve_HrR+ cohort indicated that the RFS and OS benefit of NAC was greater in the mt signature group than in the wt signature group. From post hoc analyses, the RFS and OS benefit from adding capecitabine to FEC+T as NAC might be observed only in the mt signature group. The TP53 signature can predict the pCR after NAC, and the RFS and OS benefit from NAC may be greater in the mt signature group than in the wt signature group.
Collapse
Affiliation(s)
- Shin Takahashi
- Department of Medical Oncology, Tohoku University Hospital, Sendai, Japan; Department of Clinical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Nobuaki Sato
- Department of Breast Oncology, Niigata Cancer Center Hospital, Niigata, Japan
| | - Kouji Kaneko
- Department of Breast Oncology, Niigata Cancer Center Hospital, Niigata, Japan
| | - Norikazu Masuda
- Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan; Department of Surgery, Breast Oncology, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Masaaki Kawai
- Department of Breast Surgery, Miyagi Cancer Center Hospital, Miyagi, Japan; Department of Surgery I, Yamagata University Graduate School of Medical Science, Yamagata, Japan
| | | | - Tadashi Nomizu
- Department of Surgery, Hoshi General Hospital, Fukushima, Japan
| | - Hiroji Iwata
- Department of Breast Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Ai Ueda
- Department of Breast Oncology and Surgery, Tokyo Medical University Hospital, Tokyo, Japan
| | - Takashi Ishikawa
- Department of Breast Oncology and Surgery, Tokyo Medical University Hospital, Tokyo, Japan
| | - Hiroko Bando
- Breast and Endocrine Surgery, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Yuka Inoue
- Breast Oncology Center, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takayuki Ueno
- Breast Oncology Center, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Shinji Ohno
- Breast Oncology Center, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Makoto Kubo
- Department of Breast Surgical Oncology, Kyushu University Hospital, Kyushu University, Fukuoka, Japan
| | - Hideko Yamauchi
- Department of Breast Surgical Oncology, St. Luke's International Hospital, Tokyo, Japan
| | - Masahiro Okamoto
- Department of Breast Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Eriko Tokunaga
- Department of Breast Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Shunji Kamigaki
- Department of Surgery, Sakai Municipal Hospital, Sakai, Japan
| | - Kenjiro Aogi
- Department of Breast Oncology, National Hospital Organization Shikoku Cancer Center, Ehime, Japan
| | - Hideaki Komatsu
- Department of Surgery, Iwate Medical University School of Medicine, Shiwa, Japan
| | - Masahiro Kitada
- Breast Disease Center, Asahikawa Medical University Hospital, Asahikawa, Japan
| | - Yasuaki Uemoto
- Department of Breast Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Tatsuya Toyama
- Department of Breast Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yutaka Yamamoto
- Department of Breast and Endocrine Surgery, Kumamoto University Hospital, Kumamoto, Japan
| | - Toshinari Yamashita
- Department of Breast Surgery and Oncology, Kanagawa Cancer Center, Yokohama, Japan
| | - Takehiro Yanagawa
- Department of Breast Surgery, Kansai Rosai Hospital, Amagasaki, Japan
| | - Hiroko Yamashita
- Department of Breast Surgery, Hokkaido University Hospital, Sapporo, Japan
| | - Yoshiaki Matsumoto
- Breast Cancer Unit, Kyoto University Hospital, Graduate School of Medicine, Kyoto, Japan
| | - Masakazu Toi
- Breast Cancer Unit, Kyoto University Hospital, Graduate School of Medicine, Kyoto, Japan
| | - Minoru Miyashita
- Department of Breast and Endocrine Surgical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takanori Ishida
- Department of Breast and Endocrine Surgical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | - Satoko Sato
- Department of Pathology, Tohoku University Hospital, Sendai, Japan
| | - Takuhiro Yamaguchi
- Division of Biostatistics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Fumiaki Takahashi
- Division of Medical Engineering, Department of Information Science, Iwate Medical University, Yahaba, Japan
| | - Chikashi Ishioka
- Department of Medical Oncology, Tohoku University Hospital, Sendai, Japan; Department of Clinical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| |
Collapse
|
5
|
Qureshi Z, Jamil A, Altaf F, Siddique R, Fatima E, Dost S, Zelkowitz RS, Shah S. Revolutionizing triple-negative metastatic breast cancer treatment: sacituzumab Govitecan's role in advancing chemotherapy. Ann Med Surg (Lond) 2024; 86:5314-5319. [PMID: 39238964 PMCID: PMC11374285 DOI: 10.1097/ms9.0000000000002347] [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: 04/28/2024] [Accepted: 06/25/2024] [Indexed: 09/07/2024] Open
Abstract
Purpose This review aims to discuss the role and efficacy of Sacituzumab Govitecan in the management of breast cancer. Summary Breast cancer is the most prevalent type of cancer among women worldwide. This comprehensive review delves into the advancements brought about by Sacituzumab Govitecan in the treatment of metastatic triple-negative breast cancer (TNBC). With a focus on its mode of action, efficacious role, clinical trials, and comparative advantages over conventional chemotherapy, the review highlights the therapy's precision in targeting cancer cells through monoclonal antibodies. Sacituzumab Govitecan's ability to deliver a chemotherapeutic payload specifically to cancer cells with the Trop-2 receptor sets it apart from traditional chemotherapy, minimizing collateral damage and reducing severe side effects. The impact of Sacituzumab Govitecan on improving progression-free survival, tumor response rates, and, significantly, the quality of life for patients is discussed. This article also sheds light on ongoing trials, FDA recognition, and the therapy's potential to transform breast cancer treatment. Conclusion In conclusion, Sacituzumab Govitecan shows potential as an innovative therapeutic option for breast cancer, particularly in metastatic breast cancer and triple-negative breast cancer, but it warrants additional research.
Collapse
Affiliation(s)
- Zaheer Qureshi
- The Frank H. Netter M.D. School of Medicine at Quinnipiac University
| | - Abdur Jamil
- Department of Medicine, Samaritan Medical Centre
| | - Faryal Altaf
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai/BronxCare Health System, New York, NY, USA
| | | | - Eeshal Fatima
- Department of Medicine, Services Institute of Medical Sciences, Lahore, Pakistan
| | - Sara Dost
- Department of Hematology/Oncology, St. Vincent Medical Center, Bridgeport, CT
| | | | - Shivendra Shah
- Department of Medicine, Nepalgunj Medical College, Chisapani, Nepal
| |
Collapse
|
6
|
Wu Z, Lin Q, Wang H, Chen J, Wang G, Fu G, Li L, Bian T. Intratumoral and Peritumoral Radiomics Based on Preoperative MRI for Evaluation of Programmed Cell Death Ligand-1 Expression in Breast Cancer. J Magn Reson Imaging 2024; 60:588-599. [PMID: 37916918 DOI: 10.1002/jmri.29109] [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: 08/06/2023] [Revised: 10/17/2023] [Accepted: 10/17/2023] [Indexed: 11/03/2023] Open
Abstract
BACKGROUND Programmed cell death ligand-1 (PD-L1) is a promising target for immune checkpoint blockade therapy in breast cancer. However, the preoperative evaluation of PD-L1 expression in breast cancer is rarely explored. PURPOSE To determine the ability of radiomics signatures based on preoperative dynamic contrast-enhanced (DCE) MRI to evaluate PD-L1 expression in breast cancer. STUDY TYPE Retrospective. POPULATION 196 primary breast cancer patients with preoperative MRI and postoperative pathological evaluation of PD-L1 expression, divided into training (n = 137, 28 PD-L1-positive) and test cohorts (n = 59, 12 PD-L1-positive). FIELD STRENGTH/SEQUENCE 3.0T; volume imaging for breast assessment DCE sequence. ASSESSMENT Radiomics features were extracted from the first phase of DCE-MRI by using the minimum redundancy maximum relevance method and least absolute shrinkage and selection operator algorithm. Three radiomics signatures were constructed based on the intratumoral, peritumoral, and combined intra- and peritumoral regions. The performance of the signatures was assessed using area under the receiver operating characteristic (ROC) curve (AUC), sensitivity, specificity, and accuracy. STATISTICAL TESTS Univariable and multivariable logistic regression analysis, t-tests, chi-square tests, Fisher exact test or Yates correction, ROC analysis, and one-way analysis of variance. P < 0.05 was considered significant. RESULTS In the test cohort, the combined radiomics signature (AUC, 0.853) exhibited superior performance compared to the intratumoral (AUC, 0.816; P = 0.528) and peritumoral radiomics signatures (AUC, 0.846; P = 0.905) in PD-L1 status evaluation, although the differences did not reach statistical significance. DATA CONCLUSION Intratumoral and peritumoral radiomics signatures based on preoperative breast MRI showed some potential accuracy for the non-invasive evaluation of PD-L1 status in breast cancer. LEVEL OF EVIDENCE 4 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Zengjie Wu
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Qing Lin
- Breast Disease Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Haibo Wang
- Breast Disease Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jingjing Chen
- Breast Disease Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Guanqun Wang
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Guangming Fu
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Lili Li
- Breast Disease Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Tiantian Bian
- Breast Disease Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
7
|
Dvir K, Giordano S, Leone JP. Immunotherapy in Breast Cancer. Int J Mol Sci 2024; 25:7517. [PMID: 39062758 PMCID: PMC11276856 DOI: 10.3390/ijms25147517] [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: 06/12/2024] [Revised: 06/29/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Breast cancer is a disease encompassing a spectrum of molecular subtypes and clinical presentations, each with distinct prognostic implications and treatment responses. Breast cancer has traditionally been considered an immunologically "cold" tumor, unresponsive to immunotherapy. However, clinical trials in recent years have found immunotherapy to be an efficacious therapeutic option for select patients. Breast cancer is categorized into different subtypes ranging from the most common positive hormone receptor (HR+), human epidermal growth factor receptor 2 (HER2)-negative type, to less frequent HER2- positive breast cancer and triple-negative breast cancer (TNBC), highlighting the necessity for tailored treatment strategies aimed at maximizing patient outcomes. Despite notable progress in early detection and new therapeutic modalities, breast cancer remains the second leading cause of cancer death in the USA. Moreover, in recent decades, breast cancer incidence rates have been increasing, especially in women younger than the age of 50. This has prompted the exploration of new therapeutic approaches to address this trend, offering new therapeutic prospects for breast cancer patients. Immunotherapy is a class of therapeutic agents that has revolutionized the treatment landscape of many cancers, namely melanoma, lung cancer, and gastroesophageal cancers, amongst others. Though belatedly, immunotherapy has entered the treatment armamentarium of breast cancer, with the approval of pembrolizumab in combination with chemotherapy in triple-negative breast cancer (TNBC) in the neoadjuvant and advanced settings, thereby paving the path for further research and integration of immune checkpoint inhibitors in other subtypes of breast cancer. Trials exploring various combination therapies to harness the power of immunotherapy in symbiosis with various chemotherapeutic agents are ongoing in hopes of improving response rates and prolonging survival for breast cancer patients. Biomarkers and precise patient selection for the utilization of immunotherapy remain cardinal and are currently under investigation, with some biomarkers showing promise, such as Program Death Lignat-1 (PDL-1) Combined Positive Score, Tumor Mutation Burden (TMB), and Tumor Infiltrating Lymphocytes (TILs). This review will present the current landscape of immunotherapy, particularly checkpoint inhibitors, in different types of breast cancer.
Collapse
Affiliation(s)
- Kathrin Dvir
- Dana Farber Cancer Institute, Boston, MA 02215, USA; (K.D.)
- St. Elizabeth’s Medical Center, Boston, MA 02111, USA
| | - Sara Giordano
- Dana Farber Cancer Institute, Boston, MA 02215, USA; (K.D.)
- St. Elizabeth’s Medical Center, Boston, MA 02111, USA
| | | |
Collapse
|
8
|
Wang Y, Sun Y, Lu F, Zhao X, Nie Z, Zhu F, He B. Efficacy and safety of a combination treatment of immune checkpoint inhibitors in metastatic breast cancer: a systematic review and meta-analysis. Clin Transl Oncol 2024; 26:1725-1737. [PMID: 38587602 DOI: 10.1007/s12094-024-03396-6] [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: 09/27/2023] [Accepted: 01/22/2024] [Indexed: 04/09/2024]
Abstract
PURPOSE Immune checkpoint inhibitors (ICIs) in combination with chemotherapy have showed its benefits in clinical studies, and here we conducted a further evaluation on the safety and efficacy of this treatment strategy. METHODS A systematic literature review was conducted in PubMed, Embase and Cochrane Library to identify clinical studies on ICIs and chemotherapy for metastatic breast cancer. The primary efficacy endpoints were progression-free survival (PFS) and overall survival (OS), and adverse events (AEs) were analyzed. Random or fixed effects models were used to estimate pooled Hazard ratio (HR), odds ratio (OR) and the data of 95% confidence interval (CI) depend on the Heterogeneity. Cochrane risk assessment tool was used to assess risk of bias. We also drew forest plots and funnel plots, respectively. RESULTS Seven studies with intend-to-treat (ITT) population for 3255 patients were analyzed. ICIs pooled therapy showed clinical benefits compared with chemotherapy alone, improving PFS (HR = 0.81, 95% CI: 0.74-0.90) of patients with metastatic triple negative breast cancer (mTNBC), especially in patients with PD-L1-positive tumors. However, it had no effect on OS (HR = 0.92, 95% CI 0.85-1.01). Besides, mTNBC patients received pooled therapy were less frequently to have AEs (OR = 1.30, 95% CI: 1.09-1.54). In patients with metastatic Human Epidermal Growth Factor Receptor 2 (HER2) negative breast cancer, pooled therapy showed no benefit for PFS (HR = 0.80, 95% CI: 0.50-1.28) and OS (HR = 0.87, 95% CI: 0.48-1.58). CONCLUSION Pooled therapy had improved PFS in mTNBC patients, especially in patients with PD-L1-positive tumors, and it was less likely to cause grade ≥ 3 AEs.
Collapse
Affiliation(s)
- Ying Wang
- School of Basic-Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Deparment of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Yalan Sun
- School of Basic-Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Deparment of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Fang Lu
- School of Basic-Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Deparment of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Xianghong Zhao
- School of Basic-Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Deparment of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Zhenlin Nie
- Deparment of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Feng Zhu
- Department of Laboratory Medicine, Nanjing Jiangning People's Hospital, 68 Gushan Road, Jiangning District, Nanjing, Jiangsu, 211100, China.
| | - Bangshun He
- School of Basic-Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.
- Deparment of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China.
| |
Collapse
|
9
|
Kuryk L, Mathlouthi S, Wieczorek M, Gad B, Rinner B, Malfanti A, Mastrotto F, Salmaso S, Caliceti P, Garofalo M. Priming with oncolytic adenovirus followed by anti-PD-1 and paclitaxel treatment leads to improved anti-cancer efficacy in the 3D TNBC model. Eur J Pharm Biopharm 2024; 199:114300. [PMID: 38697488 DOI: 10.1016/j.ejpb.2024.114300] [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: 01/30/2024] [Revised: 04/05/2024] [Accepted: 04/18/2024] [Indexed: 05/05/2024]
Abstract
Triple-negative breast cancer (TNBC) is considered one of the most incurable malignancies due to its clinical characteristics, including high invasiveness, high metastatic potential, proneness to relapse, and poor prognosis. Therefore, it remains a critical unmet medical need. On the other hand, poor delivery efficiency continues to reduce the efficacy of anti-cancer therapeutics developed against solid tumours using various strategies, such as genetically engineered oncolytic vectors used as nanocarriers. The study was designed to evaluate the anti-tumour efficacy of a novel combinatorial therapy based on oncolytic adenovirus AdV5/3-D24-ICOSL-CD40L with an anti-PD-1 (pembrolizumab) and paclitaxel (PTX). Here, we first tested the antineoplastic effect in two-dimensional (2D) and three-dimensional (3D) breast cancer models in MDA-MB-231, MDA-MB-468 and MCF-7 cells. Then, to further evaluate the efficacy of combinatorial therapy, including immunological aspects, we established a three-dimensional (3D) co-culture model based on MDA-MB-231 cells with peripheral blood mononuclear cells (PBMCs) to create an integrated system that more closely mimics the complexity of the tumour microenvironment and interacts with the immune system. Treatment with OV as a priming agent, followed by pembrolizumab and then paclitaxel, was the most effective in reducing the tumour volume in TNBC co-cultured spheroids. Further, T-cell phenotyping analyses revealed significantly increased infiltration of CD8+, CD4+ T and Tregs cells. Moreover, the observed anti-tumour effects positively correlated with the level of CD4+ T cell infiltrates, suggesting the development of anti-cancer immunity. Our study demonstrated that combining different immunotherapeutic agents (virus, pembrolizumab) with PTX reduced the tumour volume of the TNBC co-cultured spheroids compared to relevant controls. Importantly, sequential administration of the investigational agents (priming with the vector) further enhanced the anti-cancer efficacy in 3D culture over other groups tested. Taken together, these results support further evaluation of the virus in combination with anti-PD-1 and PTX for the treatment of triple-negative breast cancer patients. Importantly, further studies with in vivo models should be conducted to better understand the translational aspects of tested therapy.
Collapse
Affiliation(s)
- Lukasz Kuryk
- Department of Virology, National Institute of Public Health NIH - National Research Institute, Chocimska 24, 00-791 Warsaw, Poland.
| | - Sara Mathlouthi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via F. Marzolo 5, 35131 Padua, Italy
| | - Magdalena Wieczorek
- Department of Virology, National Institute of Public Health NIH - National Research Institute, Chocimska 24, 00-791 Warsaw, Poland
| | - Beata Gad
- Department of Virology, National Institute of Public Health NIH - National Research Institute, Chocimska 24, 00-791 Warsaw, Poland
| | - Beate Rinner
- Division of Biomedical Research, Medical University of Graz, Roseggerweg 48, 8036 Graz, Austria
| | - Alessio Malfanti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via F. Marzolo 5, 35131 Padua, Italy
| | - Francesca Mastrotto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via F. Marzolo 5, 35131 Padua, Italy
| | - Stefano Salmaso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via F. Marzolo 5, 35131 Padua, Italy
| | - Paolo Caliceti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via F. Marzolo 5, 35131 Padua, Italy
| | - Mariangela Garofalo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via F. Marzolo 5, 35131 Padua, Italy.
| |
Collapse
|
10
|
Abraham MJ, Goncalves C, McCallum P, Gupta V, Preston SEJ, Huang F, Chou H, Gagnon N, Johnson NA, Miller WH, Mann KK, Del Rincon SV. Tunable PhenoCycler imaging of the murine pre-clinical tumour microenvironments. Cell Biosci 2024; 14:19. [PMID: 38311785 PMCID: PMC10840224 DOI: 10.1186/s13578-024-01199-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/19/2024] [Indexed: 02/06/2024] Open
Abstract
BACKGROUND The tumour microenvironment (TME) consists of tumour-supportive immune cells, endothelial cells, and fibroblasts. PhenoCycler, a high-plex single cell spatial biology imaging platform, is used to characterize the complexity of the TME. Researchers worldwide harvest and bank tissues from mouse models which are employed to model a plethora of human disease. With the explosion of interest in spatial biology, these panoplies of archival tissues provide a valuable resource to answer new questions. Here, we describe our protocols for developing tunable PhenoCycler multiplexed imaging panels and describe our open-source data analysis pipeline. Using these protocols, we used PhenoCycler to spatially resolve the TME of 8 routinely employed pre-clinical models of lymphoma, breast cancer, and melanoma preserved as FFPE. RESULTS Our data reveal distinct TMEs in the different cancer models that were imaged and show that cell-cell contacts differ depending on the tumour type examined. For instance, we found that the immune infiltration in a murine model of melanoma is altered in cellular organization in melanomas that become resistant to αPD-1 therapy, with depletions in a number of cell-cell interactions. CONCLUSIONS This work presents a valuable resource study seamlessly adaptable to any field of research involving murine models. The methodology described allows researchers to address newly formed hypotheses using archival materials, bypassing the new to perform new mouse studies.
Collapse
Affiliation(s)
- Madelyn J Abraham
- Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada
- Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | | | - Paige McCallum
- Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada
- Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Vrinda Gupta
- Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada
- Division of Experimental Medicine, McGill University, Montreal, QC, Canada
- University of British Columbia, Vancouver, BC, Canada
| | - Samuel E J Preston
- Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada
- Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Fan Huang
- Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada
- Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Hsiang Chou
- Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada
- Clinical Research Unit, Jewish General Hospital, Montreal, QC, Canada
| | - Natascha Gagnon
- Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada
| | - Nathalie A Johnson
- Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada
- Division of Experimental Medicine, McGill University, Montreal, QC, Canada
- Clinical Research Unit, Jewish General Hospital, Montreal, QC, Canada
| | - Wilson H Miller
- Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada.
- Division of Experimental Medicine, McGill University, Montreal, QC, Canada.
- Clinical Research Unit, Jewish General Hospital, Montreal, QC, Canada.
| | - Koren K Mann
- Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada.
- Division of Experimental Medicine, McGill University, Montreal, QC, Canada.
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada.
| | - Sonia V Del Rincon
- Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada.
- Division of Experimental Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
11
|
Huang M, Fasching PA, Haiderali A, Xue W, Pan W, Karantza V, Yang F, Truscott J, Xin Y, O'Shaughnessy J. Association between event-free survival and overall survival in early-stage triple-negative breast cancer. Future Oncol 2024; 20:335-348. [PMID: 37602372 DOI: 10.2217/fon-2023-0315] [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: 08/22/2023] Open
Abstract
Aim: This study evaluated event-free survival (EFS) as a surrogate outcome for overall survival (OS) in neoadjuvant therapy for early-stage triple-negative breast cancer (eTNBC). Methods: Meta-regression analyses based on a targeted literature review were used to evaluate the individual- and trial-level associations between EFS and OS. Results: In the individual-level analyses, 3-year EFS was a significant predictor of 5-year OS (p < 0.01; coefficient of determinations [R2]: 0.82 [95% CI: 0.68-0.91]). Additionally, there was a statistically significant association between the treatment effect on EFS and OS at the trial level (p < 0.001; R2: 0.64 [95% CI: 0.45-0.82]). Conclusion: This study demonstrates significant associations between EFS and OS and suggests that EFS is a valid surrogate for OS following neoadjuvant therapy for eTNBC.
Collapse
Affiliation(s)
| | - Peter A Fasching
- Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Department of Gynecology & Obstetrics, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | | | | | | | | | | | | | | | - Joyce O'Shaughnessy
- Baylor University Medical Center, Texas Oncology & US Oncology, Dallas, TX, USA
| |
Collapse
|
12
|
Tabrizian P, Yu A, Debnath N, Myers B, Marron T. Immunotherapy and Liver Transplantation: The Future or the Failure? Surg Clin North Am 2024; 104:163-182. [PMID: 37953034 DOI: 10.1016/j.suc.2023.07.009] [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] [Indexed: 11/14/2023]
Abstract
A quarter century has passed since the milestone study by Mazzaferro and colleagues on liver transplantation (LT) for hepatocellular carcinoma (HCC). The increasing demand for LT for HCC has led to the continued efforts to expand LT indications. Downstaging to within Milan criteria has been incorporated into the organ allocation policy for HCC in the United States in 2017 and provides acceptable long-term survival. The present review focuses on the rationale of neoadjuvant immune checkpoint inhibitor (ICI) in HCC, the experience of ICI in the pre- and posttransplant setting.
Collapse
Affiliation(s)
- Parissa Tabrizian
- Liver Transplant and Hepatobiliary Surgery, Recanati/Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy, New York, NY 10029, USA.
| | - Allen Yu
- Liver Transplant and Hepatobiliary Surgery, Recanati/Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy, New York, NY 10029, USA
| | - Neha Debnath
- Liver Transplant and Hepatobiliary Surgery, Recanati/Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy, New York, NY 10029, USA
| | - Bryan Myers
- Liver Transplant and Hepatobiliary Surgery, Recanati/Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy, New York, NY 10029, USA
| | - Thomas Marron
- Liver Transplant and Hepatobiliary Surgery, Recanati/Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy, New York, NY 10029, USA
| |
Collapse
|
13
|
Wang H, Wang R, Luo L, Hong J, Chen X, Shen K, Wang Y, Huang R, Wang Z. An exosome-based specific transcriptomic signature for profiling regulation patterns and modifying tumor immune microenvironment infiltration in triple-negative breast cancer. Front Immunol 2023; 14:1295558. [PMID: 38124743 PMCID: PMC10731294 DOI: 10.3389/fimmu.2023.1295558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly heterogeneous tumor that lacks effective treatment and has a poor prognosis. Exosomes carry abundant genomic information and have a significant role in tumorigenesis, metastasis, and drug resistance. However, further exploration is needed to investigate the relationship between exosome-related genes and the heterogeneity and tumor immune microenvironment of TNBC. Based on the exosome-related gene sets, multiple machine learning algorithms, such as Cox boost, were used to screen the risk score model with the highest C-index. A 9-gene risk score model was constructed, and the TNBC population was divided into high- and low-risk groups. The effectiveness of this model was verified in multiple datasets. Compared with the low-risk group, the high-risk group exhibited a poorer prognosis, which may be related to lower levels of immune infiltration and immune response rates. The gene mutation profiles and drug sensitivity of the two groups were also compared. By screening for genes with the most prognostic value, the hub gene, CLDN7, was identified, and thus, its potential role in predicting prognosis, as well as providing ideas for the clinical diagnosis, treatment, and risk assessment of TNBC, was also discussed. This study demonstrates that exosome-related genes can be used for risk stratification in TNBC, identifying patients with a worse prognosis. The high-risk group exhibited a poorer prognosis and required more aggressive treatment strategies. Analysis of the genomic information in patient exosomes may help to develop personalized treatment decisions and improve their prognosis. CLDN7 has potential value in prognostic prediction in the TNBC population.
Collapse
Affiliation(s)
- Han Wang
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruo Wang
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Luo
- Institute of Microsurgery on Extremities, Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jin Hong
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaosong Chen
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kunwei Shen
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yang Wang
- Institute of Microsurgery on Extremities, Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Renhong Huang
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Wang
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
14
|
Schäffler H, Mergel F, Pfister K, Lukac S, Fink A, Veselinovic K, Rack B, Fink V, Leinert E, Dimpfl M, Englisch A, Tegeler CM, Seller A, Grischke EM, Hahn M, Volmer LL, Engler T, Frevert ML, Taran FA, Janni W, Brucker SY, Hartkopf AD, Dannehl D. The Clinical Relevance of the NATALEE Study: Application of the NATALEE Criteria to a Real-World Cohort from Two Large German Breast Cancer Centers. Int J Mol Sci 2023; 24:16366. [PMID: 38003555 PMCID: PMC10671738 DOI: 10.3390/ijms242216366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/31/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
The NATALEE study showed a significant benefit in invasive disease-free survival (iDFS) for patients with HR+/HER2- early breast cancer (eBC) at intermediate and high risk of recurrence who were treated with the CDK4/6 inhibitor Ribociclib in combination with endocrine therapy (ET). This retrospective study aims to apply the NATALEE inclusion criteria to a representative real-world cohort to estimate the proportion of HR+/HER2- breast cancer patients eligible for adjuvant Ribociclib therapy. Patients who underwent full surgical treatment for eBC between January 2018 and December 2020 at two large German university breast cancer centers (University of Ulm, University of Tuebingen) were included. Descriptive statistics were used to characterize the patient population eligible for Ribociclib treatment based on the NATALEE study's inclusion criteria. Out of 2384 enrolled patients, 1738 had HR+/HER2- eBC, of whom 43% (747/1738) met the NATALEE inclusion criteria. Of note, these patients were older, received less chemotherapy and presented with less advanced tumor stages compared to the NATALEE study cohort. Additionally, compared to the NATALEE study cohort, fewer patients had lymph node involvement (72.4% vs. 88.7%). Our analysis suggests that approximately 43% of all HR+/HER2- breast cancer patients will qualify for Ribociclib treatment. Given the numerous treatment options for patients with HR+/HER2- eBC, as well as the differences between the NATALEE cohort and patients in the real-world clinical setting, future analyses will be needed to determine which patients would benefit most from adjuvant CDK4/6 inhibitor treatment.
Collapse
Affiliation(s)
- Henning Schäffler
- Department of Gynecology and Obstetrics, University Hospital Ulm, 89075 Ulm, Germany; (F.M.); (A.F.); (E.L.); (W.J.)
| | - Franziska Mergel
- Department of Gynecology and Obstetrics, University Hospital Ulm, 89075 Ulm, Germany; (F.M.); (A.F.); (E.L.); (W.J.)
| | - Kerstin Pfister
- Department of Gynecology and Obstetrics, University Hospital Ulm, 89075 Ulm, Germany; (F.M.); (A.F.); (E.L.); (W.J.)
| | - Stephan Lukac
- Department of Gynecology and Obstetrics, University Hospital Ulm, 89075 Ulm, Germany; (F.M.); (A.F.); (E.L.); (W.J.)
| | - Angelina Fink
- Department of Gynecology and Obstetrics, University Hospital Ulm, 89075 Ulm, Germany; (F.M.); (A.F.); (E.L.); (W.J.)
| | - Kristina Veselinovic
- Department of Gynecology and Obstetrics, University Hospital Ulm, 89075 Ulm, Germany; (F.M.); (A.F.); (E.L.); (W.J.)
| | - Brigitte Rack
- Department of Gynecology and Obstetrics, University Hospital Ulm, 89075 Ulm, Germany; (F.M.); (A.F.); (E.L.); (W.J.)
| | - Visnja Fink
- Department of Gynecology and Obstetrics, University Hospital Ulm, 89075 Ulm, Germany; (F.M.); (A.F.); (E.L.); (W.J.)
| | - Elena Leinert
- Department of Gynecology and Obstetrics, University Hospital Ulm, 89075 Ulm, Germany; (F.M.); (A.F.); (E.L.); (W.J.)
| | - Moritz Dimpfl
- Department of Gynecology and Obstetrics, University Hospital Mannheim, 68135 Mannheim, Germany;
| | - Alexander Englisch
- Department of Women’s Health, Tuebingen University, 72076 Tuebingen, Germany; (A.E.); (C.M.T.); (A.S.); (E.-M.G.); (M.H.); (L.L.V.); (T.E.); (A.D.H.); (D.D.)
| | - Christian Martin Tegeler
- Department of Women’s Health, Tuebingen University, 72076 Tuebingen, Germany; (A.E.); (C.M.T.); (A.S.); (E.-M.G.); (M.H.); (L.L.V.); (T.E.); (A.D.H.); (D.D.)
| | - Anna Seller
- Department of Women’s Health, Tuebingen University, 72076 Tuebingen, Germany; (A.E.); (C.M.T.); (A.S.); (E.-M.G.); (M.H.); (L.L.V.); (T.E.); (A.D.H.); (D.D.)
| | - Eva-Maria Grischke
- Department of Women’s Health, Tuebingen University, 72076 Tuebingen, Germany; (A.E.); (C.M.T.); (A.S.); (E.-M.G.); (M.H.); (L.L.V.); (T.E.); (A.D.H.); (D.D.)
| | - Markus Hahn
- Department of Women’s Health, Tuebingen University, 72076 Tuebingen, Germany; (A.E.); (C.M.T.); (A.S.); (E.-M.G.); (M.H.); (L.L.V.); (T.E.); (A.D.H.); (D.D.)
| | - Léa Louise Volmer
- Department of Women’s Health, Tuebingen University, 72076 Tuebingen, Germany; (A.E.); (C.M.T.); (A.S.); (E.-M.G.); (M.H.); (L.L.V.); (T.E.); (A.D.H.); (D.D.)
| | - Tobias Engler
- Department of Women’s Health, Tuebingen University, 72076 Tuebingen, Germany; (A.E.); (C.M.T.); (A.S.); (E.-M.G.); (M.H.); (L.L.V.); (T.E.); (A.D.H.); (D.D.)
| | - Marie Louise Frevert
- Department of Obstetrics and Gynecology, University of Freiburg, 79106 Freiburg, Germany; (M.L.F.)
| | - Florin Andrei Taran
- Department of Obstetrics and Gynecology, University of Freiburg, 79106 Freiburg, Germany; (M.L.F.)
| | - Wolfgang Janni
- Department of Gynecology and Obstetrics, University Hospital Ulm, 89075 Ulm, Germany; (F.M.); (A.F.); (E.L.); (W.J.)
| | - Sara Yvonne Brucker
- Department of Women’s Health, Tuebingen University, 72076 Tuebingen, Germany; (A.E.); (C.M.T.); (A.S.); (E.-M.G.); (M.H.); (L.L.V.); (T.E.); (A.D.H.); (D.D.)
| | - Andreas Daniel Hartkopf
- Department of Gynecology and Obstetrics, University Hospital Ulm, 89075 Ulm, Germany; (F.M.); (A.F.); (E.L.); (W.J.)
- Department of Women’s Health, Tuebingen University, 72076 Tuebingen, Germany; (A.E.); (C.M.T.); (A.S.); (E.-M.G.); (M.H.); (L.L.V.); (T.E.); (A.D.H.); (D.D.)
| | - Dominik Dannehl
- Department of Women’s Health, Tuebingen University, 72076 Tuebingen, Germany; (A.E.); (C.M.T.); (A.S.); (E.-M.G.); (M.H.); (L.L.V.); (T.E.); (A.D.H.); (D.D.)
| |
Collapse
|
15
|
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
|
16
|
Divoux J, Florent R, Jacobs M, Lequesne J, Grellard JM, San C, Grossi S, Kerdja K, Clarisse B, Boudier G, Cherifi F, Briand M, Dolivet E, Johnson A, Dubois B, Harter V, Lacroix J, Raboutet C, Marie B, Rousseau N, Blanc-Fournier C, Vaur D, Figeac M, Poulain L, Weiswald LB, Emile G. The TRIPLEX study: use of patient-derived tumor organoids as an innovative tool for precision medicine in triple-negative breast cancer. BMC Cancer 2023; 23:883. [PMID: 37726786 PMCID: PMC10508010 DOI: 10.1186/s12885-023-11362-8] [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: 07/17/2023] [Accepted: 09/01/2023] [Indexed: 09/21/2023] Open
Abstract
BACKGROUND Triple negative breast cancers (TNBC) account for approximately 15% of all breast cancers and are associated with a shorter median survival mainly due to locally advanced tumor and high risk of metastasis. The current neoadjuvant treatment for TNBC consists of a regimen of immune checkpoint blocker and chemotherapy (chemo-ICB). Despite the frequent use of this combination for TNBC treatment, moderate results are observed and its clinical benefit in TNBC remains difficult to predict. Patient-derived tumor organoids (PDTO) are 3D in vitro cellular structures obtained from patient's tumor samples. More and more evidence suggest that these models could predict the response of the tumor from which they are derived. PDTO may thus be used as a tool to predict chemo-ICB efficacy in TNBC patients. METHOD The TRIPLEX study is a single-center observational study conducted to investigate the feasibility of generating PDTO from TNBC and to evaluate their ability to predict clinical response. PDTO will be obtained after the dissociation of biopsies and embedding into extra cellular matrix. PDTO will be cultured in a medium supplemented with growth factors and signal pathway inhibitors. Molecular and histological analyses will be performed on established PDTO lines to validate their phenotypic proximity with the original tumor. Response of PDTO to chemo-ICB will be assessed using co-cultures with autologous immune cells collected from patient blood samples. PDTO response will finally be compared with the response of the patient to evaluate the predictive potential of the model. DISCUSSION This study will allow to assess the feasibility of using PDTO as predictive tools for the evaluation of the response of TNBC patients to treatments. In the event that PDTO could faithfully predict patient response in clinically relevant time frames, a prospective clinical trial could be designed to use PDTO to guide clinical decision. This study will also permit the establishment of a living biobank of TNBC PDTO usable for future innovative strategies evaluation. TRIAL REGISTRATION The clinical trial (version 1.2) has been validated by local research ethic committee on December 30th 2021 and registered at ClinicalTrials.gov with the identifier NCT05404321 on June 3rd 2022, version 1.2.
Collapse
Affiliation(s)
- Jordane Divoux
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
- US PLATON, ORGAPRED Core Facility, Université de Caen Normandie, Caen, France
| | - Romane Florent
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
- US PLATON, ORGAPRED Core Facility, Université de Caen Normandie, Caen, France
| | - Margaux Jacobs
- Comprehensive Cancer Center François Baclesse, Breast Cancer Unit, UNICANCER, Institut Normand du Sein, Caen, France
| | - Justine Lequesne
- Comprehensive Cancer Center François Baclesse, Clinical Research Department, UNICANCER, Caen, France
| | - Jean-Michel Grellard
- Comprehensive Cancer Center François Baclesse, Clinical Research Department, UNICANCER, Caen, France
| | - Chankannira San
- Comprehensive Cancer Center François Baclesse, Clinical Research Department, UNICANCER, Caen, France
| | - Sara Grossi
- Comprehensive Cancer Center François Baclesse, Clinical Research Department, UNICANCER, Caen, France
| | - Katia Kerdja
- Comprehensive Cancer Center François Baclesse, Clinical Research Department, UNICANCER, Caen, France
| | - Bénédicte Clarisse
- Comprehensive Cancer Center François Baclesse, Clinical Research Department, UNICANCER, Caen, France
| | - Gwenaelle Boudier
- Comprehensive Cancer Center François Baclesse, Clinical Research Department, UNICANCER, Caen, France
| | - François Cherifi
- Comprehensive Cancer Center François Baclesse, Breast Cancer Unit, UNICANCER, Institut Normand du Sein, Caen, France
| | - Mélanie Briand
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France
- US PLATON, Biological Resource Center 'OvaRessources', Université de Caen Normandie, Caen, France
| | - Enora Dolivet
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France
- Comprehensive Cancer Center François Baclesse, Department of Surgery, UNICANCER, Caen, France
| | - Alisson Johnson
- Comprehensive Cancer Center François Baclesse, Breast Cancer Unit, UNICANCER, Institut Normand du Sein, Caen, France
- Comprehensive Cancer Center François Baclesse, Clinical Research Department, UNICANCER, Caen, France
| | - Brice Dubois
- Comprehensive Cancer Center François Baclesse, North-West Canceropole Data Center, UNICANCER, Caen, France
| | - Valentin Harter
- Comprehensive Cancer Center François Baclesse, North-West Canceropole Data Center, UNICANCER, Caen, France
| | - Joëlle Lacroix
- Comprehensive Cancer Center François Baclesse, Department of Radiology, UNICANCER, Caen, France
| | - Charlotte Raboutet
- Comprehensive Cancer Center François Baclesse, Department of Radiology, UNICANCER, Caen, France
| | - Brigitte Marie
- Comprehensive Cancer Center François Baclesse, Department of Radiology, UNICANCER, Caen, France
| | - Nathalie Rousseau
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
- Biological Resource Center 'Tumorotheque de Caen Basse-Normandie', IRCBN Institut Régional du Cancer Basse Normandie, Caen, France
| | - Cécile Blanc-Fournier
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France
- US PLATON, Biological Resource Center 'OvaRessources', Université de Caen Normandie, Caen, France
- Biological Resource Center 'Tumorotheque de Caen Basse-Normandie', IRCBN Institut Régional du Cancer Basse Normandie, Caen, France
- Comprehensive Cancer Center François Baclesse, Department of Biopathology, UNICANCER, Caen, France
| | - Dominique Vaur
- Comprehensive Cancer Center François Baclesse, Department of Cancer Biology and Genetics, UNICANCER, Caen, France
| | - Martin Figeac
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41 - UAR 2014 - PLBS, University of Lille, Lille, France
| | - Laurent Poulain
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France
- US PLATON, ORGAPRED Core Facility, Université de Caen Normandie, Caen, France
- US PLATON, Biological Resource Center 'OvaRessources', Université de Caen Normandie, Caen, France
| | - Louis-Bastien Weiswald
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France.
- Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France.
- US PLATON, ORGAPRED Core Facility, Université de Caen Normandie, Caen, France.
- INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), Comprehensive Cancer Center François Baclesse, 3 Avenue du Général Harris, BP 45026, Caen Cedex 05, 14 076, France.
| | - George Emile
- Comprehensive Cancer Center François Baclesse, Breast Cancer Unit, UNICANCER, Institut Normand du Sein, Caen, France.
- Comprehensive Cancer Center François Baclesse, Clinical Research Department, UNICANCER, Caen, France.
- Breast Cancer Unit, Comprehensive Cancer Center François Baclesse, 3 Avenue du Général Harris, BP 45026, Caen Cedex 05, 14 076, France.
| |
Collapse
|
17
|
Stecklein SR, Barlow W, Pusztai L, Timms K, Kennedy R, Logan GE, Seitz R, Badve S, Gökmen-Polar Y, Porter P, Linden H, Tripathy D, Hortobagyi GN, Godwin AK, Thompson A, Hayes DF, Sharma P. Dual Prognostic Classification of Triple-Negative Breast Cancer by DNA Damage Immune Response and Homologous Recombination Deficiency. JCO Precis Oncol 2023; 7:e2300197. [PMID: 37972336 PMCID: PMC10681491 DOI: 10.1200/po.23.00197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/12/2023] [Accepted: 09/11/2023] [Indexed: 11/19/2023] Open
Abstract
PURPOSE Triple-negative breast cancer (TNBC) is a heterogeneous disease. We previously showed that homologous recombination deficiency (HRD) and the DNA damage immune response (DDIR) signature are prognostic in TNBC. We hypothesized that these biomarkers reflect related but not completely interdependent biological processes, that their combined use would be prognostic, and that simultaneous assessment of the immunologic microenvironment and susceptibility to DNA damaging therapies might be able to identify subgroups with distinct therapeutic vulnerabilities. METHODS We analyzed the dual DDIR/HRD classification in 341 patients with TNBC treated with adjuvant anthracycline-based chemotherapy on the SWOG S9313 trial and corroborated our findings in The Cancer Genome Atlas breast cancer data set. RESULTS DDIR/HRD classification is highly prognostic in TNBC and identifies biologically and immunologically distinct subgroups. Immune-enriched DDIR+/HRD+ TNBCs have the most favorable prognosis, and DDIR+/HRD- and DDIR-/HRD+ TNBCs have favorable intermediate prognosis, despite the latter being immune-depleted. DDIR-/HRD- TNBCs have the worst prognosis and represent an internally heterogeneous group of immune-depleted chemoresistant tumors. CONCLUSION Our findings propose DDIR/HRD classification as a potentially clinically relevant approach to categorize tumors on the basis of therapeutic vulnerabilities.
Collapse
Affiliation(s)
| | | | | | | | - Richard Kennedy
- Almac Diagnostic Services, Craigavon, Northern Ireland, United Kingdom
- Patrick G Johnston Centre for Cancer Research, Queen's University of Belfast, Belfast, United Kingdom
| | - Gemma E. Logan
- Almac Diagnostic Services, Craigavon, Northern Ireland, United Kingdom
| | | | - Sunil Badve
- Emory University School of Medicine, Atlanta, GA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Gandhi S. Novel Biomarkers to Guide Immunotherapy De-Escalation in the Neoadjuvant Setting in Triple-Negative Breast Cancer. J Pers Med 2023; 13:1313. [PMID: 37763081 PMCID: PMC10532861 DOI: 10.3390/jpm13091313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Triple-negative breast cancer (TNBC) has the highest incidence of disease recurrence and distant metastases among breast cancer subtypes, leading to significant rates of morbidity and mortality [...].
Collapse
Affiliation(s)
- Shipra Gandhi
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| |
Collapse
|
19
|
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
|
20
|
Kahn AM, Golestani R, Harigopal M, Pusztai L. Intratumor spatial heterogeneity in programmed death-ligand 1 (PD-L1) protein expression in early-stage breast cancer. Breast Cancer Res Treat 2023:10.1007/s10549-023-06977-1. [PMID: 37378695 DOI: 10.1007/s10549-023-06977-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 05/08/2023] [Indexed: 06/29/2023]
Abstract
PURPOSE Programmed death-ligand 1 (PD-L1) expression is required for benefit from immune checkpoint inhibitors in metastatic triple negative breast cancer (TNBC). In contrast, in the neoadjuvant setting patients benefited regardless of PD-L1 expression. We hypothesized that, in stages II-III breast cancers, low levels of PD-L1 expression may be sufficient to confer sensitivity to therapy and focal expression could be missed by a biopsy. METHODS In this study, we examined intratumor spatial heterogeneity of PD-L1 protein expression in multiple biopsies from different regions of breast cancers in 57 primary breast tumors (n = 33 TNBC, n = 19 estrogen receptor-positive [ER-positive], n = 5 human epidermal receptor 2-positive [HER2 +]). E1L3N antibody was used to assess PD-L1 status and staining was scored using the combined positivity score (CPS) with PD-L1 positive defined as CPS ≥ 10. RESULTS Overall, 19% (11/57) of tumors were PD-L1 positive based on positivity in at least 1 biopsy. Among TNBC, PD-L1 positivity was 27% (9/33). The discordance rate, defined as the same tumor yielding PD-L1 positive and negative samples in different regions, was 16% (n = 9) in the whole study population and 23% (n = 7) in TNBC. Cohen's kappa coefficient of agreement was 0.214 for the whole study and 0.239 for TNBC, both of which falling into a non-statistically significant fair agreement range. Among all PD-L1 positive cases, 82% (n = 9/11) had positivity in only one of the tissue assessments. CONCLUSION These results indicate that the overall 84% concordance is driven by concordant negative results. In PD-L1 positive cancers, within-tumor heterogeneity in PD-L1 expression exists.
Collapse
Affiliation(s)
- Adriana Matutino Kahn
- Section of Medical Oncology, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
| | - Reza Golestani
- Department of Pathology, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
| | - Malini Harigopal
- Department of Pathology, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA
| | - Lajos Pusztai
- Section of Medical Oncology, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA.
- Breast Medical Oncology, Yale Cancer Center, Yale School of Medicine, 300 George St, Suite 120, Rm 133, New Haven, CT, 06520, USA.
| |
Collapse
|
21
|
Perez-Lanzon M, Carbonnier V, Cordier P, De Palma FDE, Petrazzuolo A, Klein C, Arbaretaz F, Mangane K, Stoll G, Martins I, Fohrer Ting H, Paillet J, Mouillet-Richard S, Le Corre D, Xiao W, Sroussi M, Desdouets C, Laurent-Puig P, Pol J, Lopez-Otin C, Maiuri MC, Kroemer G. New hormone receptor-positive breast cancer mouse cell line mimicking the immune microenvironment of anti-PD-1 resistant mammary carcinoma. J Immunother Cancer 2023; 11:e007117. [PMID: 37344100 PMCID: PMC10314679 DOI: 10.1136/jitc-2023-007117] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2023] [Indexed: 06/23/2023] Open
Abstract
BACKGROUND Progress in breast cancer (BC) research relies on the availability of suitable cell lines that can be implanted in immunocompetent laboratory mice. The best studied mouse strain, C57BL/6, is also the only one for which multiple genetic variants are available to facilitate the exploration of the cancer-immunity dialog. Driven by the fact that no hormone receptor-positive (HR+) C57BL/6-derived mammary carcinoma cell lines are available, we decided to establish such cell lines. METHODS BC was induced in female C57BL/6 mice using a synthetic progesterone analog (medroxyprogesterone acetate, MPA) combined with a DNA damaging agent (7,12-dimethylbenz[a]anthracene, DMBA). Cell lines were established from these tumors and selected for dual (estrogen+progesterone) receptor positivity, as well as transplantability into C57BL/6 immunocompetent females. RESULTS One cell line, which we called B6BC, fulfilled these criteria and allowed for the establishment of invasive estrogen receptor-positive (ER+) tumors with features of epithelial to mesenchymal transition that were abundantly infiltrated by myeloid immune populations but scarcely by T lymphocytes, as determined by single-nucleus RNA sequencing and high-dimensional leukocyte profiling. Such tumors failed to respond to programmed cell death-1 (PD-1) blockade, but reduced their growth on treatment with ER antagonists, as well as with anthracycline-based chemotherapy, which was not influenced by T-cell depletion. Moreover, B6BC-derived tumors reduced their growth on CD11b blockade, indicating tumor sustainment by myeloid cells. The immune environment and treatment responses recapitulated by B6BC-derived tumors diverged from those of ER+ TS/A cell-derived tumors in BALB/C mice, and of ER- E0771 cell-derived and MPA/DMBA-induced tumors in C57BL/6 mice. CONCLUSIONS B6BC is the first transplantable HR+ BC cell line derived from C57BL/6 mice and B6BC-derived tumors recapitulate the complex tumor microenvironment of locally advanced HR+ BC naturally resistant to PD-1 immunotherapy.
Collapse
Affiliation(s)
- Maria Perez-Lanzon
- Team "Metabolism, Cancer & Immunity", Centre de Recherche des Cordeliers, UMRS 1138, Inserm, Université Paris Cité, Sorbonne Université, Paris, France
- Gustave Roussy Institute, Villejuif, France
| | - Vincent Carbonnier
- Team "Metabolism, Cancer & Immunity", Centre de Recherche des Cordeliers, UMRS 1138, Inserm, Université Paris Cité, Sorbonne Université, Paris, France
- Gustave Roussy Institute, Villejuif, France
| | - Pierre Cordier
- Team 'Proliferation, Stress and Liver Physiopathology', Centre de Recherche des Cordeliers, Paris, France
| | - Fatima Domenica Elisa De Palma
- Team "Metabolism, Cancer & Immunity", Centre de Recherche des Cordeliers, UMRS 1138, Inserm, Université Paris Cité, Sorbonne Université, Paris, France
- Gustave Roussy Institute, Villejuif, France
- Department of Molecular Medicine and Medical Biotechnologies, University of Napoli Federico II, Napoli, Italy
| | - Adriana Petrazzuolo
- Team "Metabolism, Cancer & Immunity", Centre de Recherche des Cordeliers, UMRS 1138, Inserm, Université Paris Cité, Sorbonne Université, Paris, France
- Gustave Roussy Institute, Villejuif, France
| | - Christophe Klein
- Centre d'Histologie, d'Imagerie cellulaire et de Cytométrie (CHIC), Centre de Recherche des Cordeliers, Paris, France, UMRS1138, Inserm, Université Paris Cité, Sorbonne Université, Paris, France
| | - Floriane Arbaretaz
- Centre d'Histologie, d'Imagerie cellulaire et de Cytométrie (CHIC), Centre de Recherche des Cordeliers, Paris, France, UMRS1138, Inserm, Université Paris Cité, Sorbonne Université, Paris, France
| | - Khady Mangane
- Team "Metabolism, Cancer & Immunity", Centre de Recherche des Cordeliers, UMRS 1138, Inserm, Université Paris Cité, Sorbonne Université, Paris, France
- Gustave Roussy Institute, Villejuif, France
| | - Gautier Stoll
- Team "Metabolism, Cancer & Immunity", Centre de Recherche des Cordeliers, UMRS 1138, Inserm, Université Paris Cité, Sorbonne Université, Paris, France
- Gustave Roussy Institute, Villejuif, France
| | - Isabelle Martins
- Team "Metabolism, Cancer & Immunity", Centre de Recherche des Cordeliers, UMRS 1138, Inserm, Université Paris Cité, Sorbonne Université, Paris, France
- Gustave Roussy Institute, Villejuif, France
| | - Helene Fohrer Ting
- Centre d'Histologie, d'Imagerie cellulaire et de Cytométrie (CHIC), Centre de Recherche des Cordeliers, Paris, France, UMRS1138, Inserm, Université Paris Cité, Sorbonne Université, Paris, France
| | - Juliette Paillet
- Team "Metabolism, Cancer & Immunity", Centre de Recherche des Cordeliers, UMRS 1138, Inserm, Université Paris Cité, Sorbonne Université, Paris, France
- Gustave Roussy Institute, Villejuif, France
| | - Sophie Mouillet-Richard
- Team 'Personalized medicine, pharmacogenomics, therapeutic optimization', Centre de Recherche des Cordeliers, Paris, France
| | - Delphine Le Corre
- Team 'Personalized medicine, pharmacogenomics, therapeutic optimization', Centre de Recherche des Cordeliers, Paris, France
| | - Wenjjin Xiao
- Team 'Personalized medicine, pharmacogenomics, therapeutic optimization', Centre de Recherche des Cordeliers, Paris, France
| | - Marine Sroussi
- Team 'Personalized medicine, pharmacogenomics, therapeutic optimization', Centre de Recherche des Cordeliers, Paris, France
| | - Chantal Desdouets
- Team 'Proliferation, Stress and Liver Physiopathology', Centre de Recherche des Cordeliers, Paris, France
| | - Pierre Laurent-Puig
- Team 'Personalized medicine, pharmacogenomics, therapeutic optimization', Centre de Recherche des Cordeliers, Paris, France
- Institut du Cancer Paris CARPEM, Institut Universitaire de France, Hôpital Européen Georges Pompidou, France-HP, Paris, France
| | - Jonathan Pol
- Team "Metabolism, Cancer & Immunity", Centre de Recherche des Cordeliers, UMRS 1138, Inserm, Université Paris Cité, Sorbonne Université, Paris, France
- Gustave Roussy Institute, Villejuif, France
| | - Carlos Lopez-Otin
- Departamento de Bioquimica y Biologia Molecular, Instituto Universitario de Oncologia (IUOPA), University of Oviedo, Oviedo, Spain
| | - Maria Chiara Maiuri
- Team "Metabolism, Cancer & Immunity", Centre de Recherche des Cordeliers, UMRS 1138, Inserm, Université Paris Cité, Sorbonne Université, Paris, France
- Gustave Roussy Institute, Villejuif, France
- Department of Molecular Medicine and Medical Biotechnologies, University of Napoli Federico II, Napoli, Italy
| | - Guido Kroemer
- Team "Metabolism, Cancer & Immunity", Centre de Recherche des Cordeliers, UMRS 1138, Inserm, Université Paris Cité, Sorbonne Université, Paris, France
- Gustave Roussy Institute, Villejuif, France
- Institut du Cancer Paris CARPEM, Institut Universitaire de France, Hôpital Européen Georges Pompidou, France-HP, Paris, France
| |
Collapse
|
22
|
Jacobs F, Agostinetto E, Miggiano C, De Sanctis R, Zambelli A, Santoro A. Hope and Hype around Immunotherapy in Triple-Negative Breast Cancer. Cancers (Basel) 2023; 15:cancers15112933. [PMID: 37296893 DOI: 10.3390/cancers15112933] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
Triple-negative breast cancer (TNBC) holds a poor prognosis compared to other breast cancer subtypes, and the development of new effective treatment strategies is an unmet medical need. TNBC has traditionally been considered not amenable to treatment with targeted agents due to a lack of actionable targets. Therefore, chemotherapy has remained the mainstay of systemic treatment for many decades. The advent of immunotherapy raised very hopeful expectations in TNBC, possibly due to higher levels of tumor-infiltrating lymphocytes, PD-L1 expression and tumor mutational burden compared to other breast cancer subtypes, that predict an effective anti-tumor immune-engagement. The results of clinical trials testing immunotherapy in TNBC led to the approval of the combination of immune checkpoint inhibitors and chemotherapy in both early and advanced settings. However, some open questions about the use of immunotherapy in TNBC still exist. These include a deeper understanding of the heterogeneity of the disease, identification of reliable predictive biomarkers of response, determination of the most appropriate chemotherapy backbone and appropriate management of potential long-term immune-related adverse events. In this review we aim to examine the available evidence on the use of immunotherapy strategies in both early and advanced TNBC, to critically discuss some of the limitations encountered in clinical research and to summarize data on novel promising immunotherapeutic strategies beyond PD-(L)1 blockade that have been investigated in the most recent trials.
Collapse
Affiliation(s)
- Flavia Jacobs
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, MI, Italy
- Academic Trials Promoting Team, Institut Jules Bordet, L'Université Libre de Bruxelles (U.L.B.), 1070 Brussels, Belgium
| | - Elisa Agostinetto
- Academic Trials Promoting Team, Institut Jules Bordet, L'Université Libre de Bruxelles (U.L.B.), 1070 Brussels, Belgium
| | - Chiara Miggiano
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, MI, Italy
- IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Via Manzoni 56, 20090 Pieve Emanuele, MI, Italy
| | - Rita De Sanctis
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, MI, Italy
- IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Via Manzoni 56, 20090 Pieve Emanuele, MI, Italy
| | - Alberto Zambelli
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, MI, Italy
- IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Via Manzoni 56, 20090 Pieve Emanuele, MI, Italy
| | - Armando Santoro
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, MI, Italy
- IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Via Manzoni 56, 20090 Pieve Emanuele, MI, Italy
| |
Collapse
|
23
|
Devan AR, Nair B, Aryan MK, Liju VB, Koshy JJ, Mathew B, Valsan A, Kim H, Nath LR. Decoding Immune Signature to Detect the Risk for Early-Stage HCC Recurrence. Cancers (Basel) 2023; 15:2729. [PMID: 37345066 PMCID: PMC10216348 DOI: 10.3390/cancers15102729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 05/02/2023] [Accepted: 05/09/2023] [Indexed: 06/23/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is often recognized as an inflammation-linked cancer, which possesses an immunosuppressive tumor microenvironment. Curative treatments such as surgical resection, liver transplantation, and percutaneous ablation are mainly applicable in the early stage and demonstrate significant improvement of survival rate in most patients. However, 70-80% of patients report HCC recurrence within 5 years of curative treatment, representing an important clinical issue. However, there is no effective recurrence marker after surgical and locoregional therapies, thus, tumor size, number, and histological features such as cancer cell differentiation are often considered as risk factors for HCC recurrence. Host immunity plays a critical role in regulating carcinogenesis, and the immune microenvironment characterized by its composition, functional status, and density undergoes significant alterations in each stage of cancer progression. Recent studies reported that analysis of immune contexture could yield valuable information regarding the treatment response, prognosis and recurrence. This review emphasizes the prognostic value of tumors associated with immune factors in HCC recurrence after curative treatment. In particular, we review the immune landscape and immunological factors contributing to early-stage HCC recurrence, and discuss the immunotherapeutic interventions to prevent tumor recurrence following curative treatments.
Collapse
Affiliation(s)
- Aswathy R. Devan
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi 682041, Kerala, India; (A.R.D.); (B.N.); (J.J.K.)
| | - Bhagyalakshmi Nair
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi 682041, Kerala, India; (A.R.D.); (B.N.); (J.J.K.)
| | | | - Vijayastelar B. Liju
- The Shraga Segal Department of Microbiology-Immunology and Genetics, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva 84105, Israel;
| | - Joel Joy Koshy
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi 682041, Kerala, India; (A.R.D.); (B.N.); (J.J.K.)
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi 682041, Kerala, India;
| | - Arun Valsan
- Department of Gastroenterology and Epatology, Amrita Institute of Medical Science, Kochi 682041, Kerala, India;
| | - Hoon Kim
- Department of Pharmacy, and Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea
| | - Lekshmi R. Nath
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi 682041, Kerala, India; (A.R.D.); (B.N.); (J.J.K.)
| |
Collapse
|
24
|
Vaz SC, Graff SL, Ferreira AR, Debiasi M, de Geus-Oei LF. PET/CT in Patients with Breast Cancer Treated with Immunotherapy. Cancers (Basel) 2023; 15:cancers15092620. [PMID: 37174086 PMCID: PMC10177398 DOI: 10.3390/cancers15092620] [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: 03/29/2023] [Revised: 04/27/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023] Open
Abstract
Significant advances in breast cancer (BC) treatment have been made in the last decade, including the use of immunotherapy and, in particular, immune checkpoint inhibitors that have been shown to improve the survival of patients with triple negative BC. This narrative review summarizes the studies supporting the use of immunotherapy in BC. Furthermore, the usefulness of 2-deoxy-2-[18F]fluoro-D-glucose (2-[18F]FDG) positron emission/computerized tomography (PET/CT) to image the tumor heterogeneity and to assess treatment response is explored, including the different criteria to interpret 2-[18F]FDG PET/CT imaging. The concept of immuno-PET is also described, by explaining the advantages of mapping treatment targets with a non-invasive and whole-body tool. Several radiopharmaceuticals in the preclinical phase are referred too, and, considering their promising results, translation to human studies is needed to support their use in clinical practice. Overall, this is an evolving field in BC treatment, despite PET imaging developments, the future trends also include expanding immunotherapy to early-stage BC and using other biomarkers.
Collapse
Affiliation(s)
- Sofia C Vaz
- Nuclear Medicine-Radiopharmacology, Champalimaud Center for the Unkown, Champalimaud Foundation, 1400-038 Lisbon, Portugal
- Department of Radiology, Leiden University Medical Center, P.O. Box 9600-2300 RC Leiden, The Netherlands
| | - Stephanie L Graff
- Division of Hematology/Oncology, Lifespan Cancer Institute, Providence, RI 02903, USA
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Arlindo R Ferreira
- Católica Medical School, Universidade Católica Portuguesa, 2635-631 Lisbon, Portugal
| | - Márcio Debiasi
- Breast Cancer Unit, Champalimaud Center for the Unkown, Champalimaud Foundation, 1400-038 Lisbon, Portugal
| | - Lioe-Fee de Geus-Oei
- Department of Radiology, Leiden University Medical Center, P.O. Box 9600-2300 RC Leiden, The Netherlands
- Biomedical Photonic Imaging Group, University of Twente, P.O. Box 217-7500 AE Enschede, The Netherlands
- Department of radiation Science & Technology, Delft University of Technology, P.O. Postbus 5 2600 AA Delft, The Netherlands
| |
Collapse
|
25
|
Desai NV, Tan AR. Targeted Therapies and the Evolving Standard of Care for Triple-Negative and Germline BRCA1/ 2-Mutated Breast Cancers in the High-Risk, Early-Stage Setting. JCO Precis Oncol 2023; 7:e2200446. [PMID: 37163718 DOI: 10.1200/po.22.00446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023] Open
Abstract
PURPOSE Immune checkpoint inhibitors (ICIs) and poly (ADP-ribose) polymerase (PARP) inhibitors have transformed the treatment landscape of metastatic triple-negative breast cancer (TNBC). Trial results have demonstrated the clinical benefit of these targeted agents in the advanced TNBC setting and have led to their evaluation in the treatment of high-risk, early-stage TNBC and BRCA-mutated breast cancer. We provide a summary of the results that have led to the establishment of the ICI pembrolizumab and the PARP inhibitor olaparib as new standards of care. METHODS Using PubMed, we searched for original articles published in English between 2017 and 2022. Search terms included triple-negative breast cancer, adjuvant, neoadjuvant, immunotherapy, and PARP inhibitors. RESULTS Two targeted therapies have been approved by the US Food and Drug Administration for the treatment of TNBC and BRCA-mutated breast cancers in the high-risk, early-stage setting on the basis of clinical trial results demonstrating improved clinical outcomes. For high-risk, early-stage TNBC, pembrolizumab was approved as neoadjuvant therapy in combination with chemotherapy and as a single agent for continued treatment after surgery; this approval was based on results of the KEYNOTE-522 trial. Olaparib was approved for the adjuvant treatment of patients with high-risk, early-stage human epidermal growth factor receptor type 2 (HER2)-negative breast cancer with germline BRCA1/2 mutations who have been previously treated with neoadjuvant or adjuvant chemotherapy on the basis of the OlympiA trial results. CONCLUSION Clinical trial results demonstrate the pronounced clinical benefits of pembrolizumab combined with chemotherapy for high-risk, early-stage TNBC and adjuvant olaparib for high-risk, early-stage HER2-negative BRCA1/2-mutated breast cancer.
Collapse
Affiliation(s)
- Neelam V Desai
- Department of Solid Tumor Oncology and Investigational Therapeutics, Levine Cancer Institute, Atrium Health, Charlotte, NC
| | - Antoinette R Tan
- Department of Solid Tumor Oncology and Investigational Therapeutics, Levine Cancer Institute, Atrium Health, Charlotte, NC
| |
Collapse
|
26
|
Geurts V, Kok M. Immunotherapy for Metastatic Triple Negative Breast Cancer: Current Paradigm and Future Approaches. Curr Treat Options Oncol 2023; 24:628-643. [PMID: 37079257 PMCID: PMC10172210 DOI: 10.1007/s11864-023-01069-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2023] [Indexed: 04/21/2023]
Abstract
OPINION STATEMENT In approximately 15-20% of the patients diagnosed with breast cancer, it comprises the triple negative (TN) subtype, which until recently lacked targets for specific treatments and is known for its aggressive clinical behavior in patients with metastatic disease. TNBC is considered the most immunogenic breast cancer subtype due to higher levels of tumor infiltrating lymphocytes (TILs), tumor mutational burden and PD-L1 expression, providing a rationale for immunotherapy. The addition of pembrolizumab to chemotherapy as first-line treatment resulted in significantly improved PFS and OS for PD-L1 positive mTNBC, leading to FDA approval. However, response rate of ICB in unselected patients is low. Ongoing (pre)clinical trials aim to further optimize ICB efficacy and widen its application beyond PD-L1 positive breast tumors. Novel immunomodulatory approaches to induce a more inflamed tumor microenvironment include dual checkpoint blockade, bispecific antibodies, immunocytokines, adoptive cell therapies, oncolytic viruses, and cancer vaccines. Preclinical data for these novel strategies seems promising, but solid clinical data to further support its application for mTNBC is awaited. Biomarkers capturing the degree of immunogenicity such as but not limited to TILs, CD8 T cell levels, and IFNg signatures could support deciding which therapeutic strategy is most appropriate for which patient. Given 1) the accumulating therapy options for patients with metastatic disease and 2) the heterogeneity of mTNBC from inflamed to immune-desert tumors, the challenge is to work towards immunomodulatory strategies for specific subgroups of patients with TNBC to enable personalized (immuno)therapy for patients with metastatic disease.
Collapse
Affiliation(s)
- Veerle Geurts
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, Plesmanlaan 121, 1066, CX, Amsterdam, The Netherlands
| | - Marleen Kok
- Division of Tumor Biology & Immunology, Netherlands Cancer Institute, Plesmanlaan 121, 1066, CX, Amsterdam, The Netherlands.
- Department of Medical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066, CX, Amsterdam, The Netherlands.
| |
Collapse
|
27
|
Yam C, Mittendorf EA, Garber HR, Sun R, Damodaran S, Murthy RK, Ramirez D, Karuturi M, Layman RM, Ibrahim N, Rauch GM, Adrada BE, Candelaria RP, White JB, Ravenberg E, Clayborn A, Ding QQ, Symmans WF, Prabhakaran S, Thompson AM, Valero V, Tripathy D, Huo L, Moulder SL, Litton JK. A phase II study of neoadjuvant atezolizumab and nab-paclitaxel in patients with anthracycline-resistant early-stage triple-negative breast cancer. Breast Cancer Res Treat 2023; 199:457-469. [PMID: 37061619 DOI: 10.1007/s10549-023-06929-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/30/2023] [Indexed: 04/17/2023]
Abstract
PURPOSE Neoadjuvant anti-PD-(L)1 therapy improves the pathological complete response (pCR) rate in unselected triple-negative breast cancer (TNBC). Given the potential for long-term morbidity from immune-related adverse events (irAEs), optimizing the risk-benefit ratio for these agents in the curative neoadjuvant setting is important. Suboptimal clinical response to initial neoadjuvant therapy (NAT) is associated with low rates of pCR (2-5%) and may define a patient selection strategy for neoadjuvant immune checkpoint blockade. We conducted a single-arm phase II study of atezolizumab and nab-paclitaxel as the second phase of NAT in patients with doxorubicin and cyclophosphamide (AC)-resistant TNBC (NCT02530489). METHODS Patients with stage I-III, AC-resistant TNBC, defined as disease progression or a < 80% reduction in tumor volume after 4 cycles of AC, were eligible. Patients received atezolizumab (1200 mg IV, Q3weeks × 4) and nab-paclitaxel (100 mg/m2 IV,Q1 week × 12) as the second phase of NAT before undergoing surgery followed by adjuvant atezolizumab (1200 mg IV, Q3 weeks, × 4). A two-stage Gehan-type design was employed to detect an improvement in pCR/residual cancer burden class I (RCB-I) rate from 5 to 20%. RESULTS From 2/15/2016 through 1/29/2021, 37 patients with AC-resistant TNBC were enrolled. The pCR/RCB-I rate was 46%. No new safety signals were observed. Seven patients (19%) discontinued atezolizumab due to irAEs. CONCLUSION This study met its primary endpoint, demonstrating a promising signal of activity in this high-risk population (pCR/RCB-I = 46% vs 5% in historical controls), suggesting that a response-adapted approach to the utilization of neoadjuvant immunotherapy should be considered for further evaluation in a randomized clinical trial.
Collapse
Affiliation(s)
- Clinton Yam
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Dan L. Duncan Building (CPB5.3542), 1515 Holcombe Blvd. Unit 1354, Houston, TX, 77030, USA.
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 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
| | - Haven R Garber
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Dan L. Duncan Building (CPB5.3542), 1515 Holcombe Blvd. Unit 1354, Houston, TX, 77030, USA
| | - Ryan Sun
- Department of Biostatistics, Division of Basic Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Senthil Damodaran
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Dan L. Duncan Building (CPB5.3542), 1515 Holcombe Blvd. Unit 1354, Houston, TX, 77030, USA
| | - Rashmi K Murthy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Dan L. Duncan Building (CPB5.3542), 1515 Holcombe Blvd. Unit 1354, Houston, TX, 77030, USA
| | - David Ramirez
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Dan L. Duncan Building (CPB5.3542), 1515 Holcombe Blvd. Unit 1354, Houston, TX, 77030, USA
| | - Meghan Karuturi
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Dan L. Duncan Building (CPB5.3542), 1515 Holcombe Blvd. Unit 1354, Houston, TX, 77030, USA
| | - Rachel M Layman
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Dan L. Duncan Building (CPB5.3542), 1515 Holcombe Blvd. Unit 1354, Houston, TX, 77030, USA
| | - Nuhad Ibrahim
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Dan L. Duncan Building (CPB5.3542), 1515 Holcombe Blvd. Unit 1354, Houston, TX, 77030, USA
| | - Gaiane M Rauch
- Department of Breast Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Beatriz E Adrada
- Department of Breast Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rosalind P Candelaria
- Department of Breast Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jason B White
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Dan L. Duncan Building (CPB5.3542), 1515 Holcombe Blvd. Unit 1354, Houston, TX, 77030, USA
| | - Elizabeth Ravenberg
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Dan L. Duncan Building (CPB5.3542), 1515 Holcombe Blvd. Unit 1354, Houston, TX, 77030, USA
| | - Alyson Clayborn
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Dan L. Duncan Building (CPB5.3542), 1515 Holcombe Blvd. Unit 1354, Houston, TX, 77030, USA
| | - Qing Qing Ding
- Department of Pathology, Division of Pathology-Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - W Fraser Symmans
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sabitha Prabhakaran
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alastair M Thompson
- Section of Breast Surgery, Division of Surgical Oncology, Baylor College of Medicine, Houston, TX, USA
| | - Vicente Valero
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Dan L. Duncan Building (CPB5.3542), 1515 Holcombe Blvd. Unit 1354, Houston, TX, 77030, USA
| | - Debu Tripathy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Dan L. Duncan Building (CPB5.3542), 1515 Holcombe Blvd. Unit 1354, Houston, TX, 77030, USA
| | - Lei Huo
- Department of Pathology, Division of Pathology-Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stacy L Moulder
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Dan L. Duncan Building (CPB5.3542), 1515 Holcombe Blvd. Unit 1354, Houston, TX, 77030, USA
| | - Jennifer K Litton
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Dan L. Duncan Building (CPB5.3542), 1515 Holcombe Blvd. Unit 1354, Houston, TX, 77030, USA.
| |
Collapse
|
28
|
Subhan MA, Parveen F, Shah H, Yalamarty SSK, Ataide JA, Torchilin VP. Recent Advances with Precision Medicine Treatment for Breast Cancer including Triple-Negative Sub-Type. Cancers (Basel) 2023; 15:2204. [PMID: 37190133 PMCID: PMC10137302 DOI: 10.3390/cancers15082204] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/04/2023] [Accepted: 04/05/2023] [Indexed: 05/17/2023] Open
Abstract
Breast cancer is a heterogeneous disease with different molecular subtypes. Breast cancer is the second leading cause of mortality in woman due to rapid metastasis and disease recurrence. Precision medicine remains an essential source to lower the off-target toxicities of chemotherapeutic agents and maximize the patient benefits. This is a crucial approach for a more effective treatment and prevention of disease. Precision-medicine methods are based on the selection of suitable biomarkers to envision the effectiveness of targeted therapy in a specific group of patients. Several druggable mutations have been identified in breast cancer patients. Current improvements in omics technologies have focused on more precise strategies for precision therapy. The development of next-generation sequencing technologies has raised hopes for precision-medicine treatment strategies in breast cancer (BC) and triple-negative breast cancer (TNBC). Targeted therapies utilizing immune checkpoint inhibitors (ICIs), epidermal growth factor receptor inhibitor (EGFRi), poly(ADP-ribose) polymerase inhibitor (PARPi), antibody-drug conjugates (ADCs), oncolytic viruses (OVs), glucose transporter-1 inhibitor (GLUT1i), and targeting signaling pathways are potential treatment approaches for BC and TNBC. This review emphasizes the recent progress made with the precision-medicine therapy of metastatic breast cancer and TNBC.
Collapse
Affiliation(s)
- Md Abdus Subhan
- Department of Chemistry, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Farzana Parveen
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
- Department of Pharmacy Services, DHQ Hospital Jhang 35200, Primary and Secondary Healthcare Department, Government of Punjab, Lahore 54000, Pakistan
| | - Hassan Shah
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
- CPBN, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | | | - Janaína Artem Ataide
- CPBN, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
- Faculty of Pharmaceutical Sciences, University of Campinas, Campinas 13083-871, SP, Brazil
| | - Valdimir P. Torchilin
- CPBN, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
29
|
Immune Checkpoint Inhibitors in Breast Cancer: A Narrative Review. Oncol Ther 2023:10.1007/s40487-023-00224-9. [PMID: 36917399 DOI: 10.1007/s40487-023-00224-9] [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: 12/20/2022] [Accepted: 02/24/2023] [Indexed: 03/16/2023] Open
Abstract
Breast cancer is the most frequently diagnosed malignancy in patients worldwide and the main cause of cancer-related death. Though still incurable, metastatic breast cancer's prognosis has been considerably improved in the past 10 years due to the introduction of new targeted agents, such as immune checkpoint inhibitors (ICI). However, these medications are associated with unique side effects known as immune-mediated adverse events (irAE). In this paper, we review the clinical evidence for the use of ICIs in breast cancer, in both the metastatic as well as neoadjuvant/adjuvant setting, followed by a review of irAE most commonly seen, and the medications used to treat them. Our opinion is that any cancer specialist treating patients with breast cancer should be aware of these side effects for early detection and management, and oncologists should be the leaders of the multidisciplinary team that will take care of them.
Collapse
|
30
|
Hoffmann O, Wormland S, Bittner AK, Collenburg M, Horn PA, Kimmig R, Kasimir-Bauer S, Rebmann V. Programmed death receptor ligand-2 (PD-L2) bearing extracellular vesicles as a new biomarker to identify early triple-negative breast cancer patients at high risk for relapse. J Cancer Res Clin Oncol 2023; 149:1159-1174. [PMID: 35366112 PMCID: PMC9984327 DOI: 10.1007/s00432-022-03980-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 03/09/2022] [Indexed: 12/24/2022]
Abstract
PURPOSE Based on the tumor-promoting features of extracellular vesicles (EV) and PD-L1/2-bearing EV subpopulations (PD-L1/2EV), we evaluated their potential as surrogate markers for disease progression or eligibility criteria for PD-1 immune checkpoint inhibition (ICI) approaches in early triple-negative breast cancer (TNBC). METHODS After enrichment of EV from plasma samples of 56 patients before and 50 after chemotherapy (CT), we determined levels of EV particle number and PD-L1/2EV by nanoparticle tracking analysis or ELISA and associated the results with clinical status/outcome and the presence of distinct circulating tumor cells (CTC) subpopulations. RESULTS Compared to healthy controls, patients had a tenfold higher EV concentration and significantly elevated PD L2EV but not PD L1EV levels. The most important clinical implications were found for PD-L2EV. High PD-L2EV levels were associated with a significantly reduced 3-year progression-free and overall survival (PFS and OS). A loss of PD-L2EV after CT was significantly more prominent in patients achieving pathological complete response (pCR). Increased pre-CT PD-L2EV levels were found in patients having NOTCH1-positive or ERBB3-positive CTC. The presence of ERBB3-positive CTC combined with high pre-CT PD-L2EV resulted in a shorter PFS. CONCLUSION This study highlights PD L2EV as a promising biomarker for risk assessment of TNBC patients and represents the basic for additional studies introducing PD-L2EV as an eligibility criterion for PD-1 ICI approaches.
Collapse
Affiliation(s)
- Oliver Hoffmann
- Department of Gynecology and Obstetrics, University Hospital of Essen, Hufelandstrasse 55, 45122, Essen, Germany.
| | - Sebastian Wormland
- Institute for Transfusion Medicine, University Hospital Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Ann-Kathrin Bittner
- Department of Gynecology and Obstetrics, University Hospital of Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Monika Collenburg
- Institute for Transfusion Medicine, University Hospital Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Peter A Horn
- Institute for Transfusion Medicine, University Hospital Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Rainer Kimmig
- Department of Gynecology and Obstetrics, University Hospital of Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Sabine Kasimir-Bauer
- Department of Gynecology and Obstetrics, University Hospital of Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Vera Rebmann
- Institute for Transfusion Medicine, University Hospital Essen, Hufelandstrasse 55, 45122, Essen, Germany.
| |
Collapse
|
31
|
Optimizing treatment for HER2-positive HR-positive breast cancer. Cancer Treat Rev 2023; 115:102529. [PMID: 36921556 DOI: 10.1016/j.ctrv.2023.102529] [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: 02/08/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023]
Abstract
Triple-positive breast tumors overexpress human epidermal growth factor receptor 2 (HER2) and are positive for hormone receptor (HR) expression. Data from real-life and clinical trials show that estrogen receptor (ER) expression affects the response to combinations of anti-HER2 and associated systemic therapies. Despite triple-positive tumors having decreased response rates compared to HR-negative/HER2-positive breast cancers, optimizing anti-HER2 treatment with dual anti-HER2 blockade remains important for optimal disease control. Preclinical data on the cross-talk between ER and growth factor receptor pathways show the efficacy of combinations of endocrine therapy and anti-HER2 drugs, which is confirmed in the clinic. Molecular dissection of triple-positive breast cancer might provide the rational for additional therapeutic strategies and the identification of promising biomarkers. This review summarizes data on systemic treatment efficacy from major clinical trials and perspectives for future clinical research in triple-positive breast cancer.
Collapse
|
32
|
Laschtowitz A, Roderburg C, Tacke F, Mohr R. Preoperative Immunotherapy in Hepatocellular Carcinoma: Current State of the Art. J Hepatocell Carcinoma 2023; 10:181-191. [PMID: 36789252 PMCID: PMC9922501 DOI: 10.2147/jhc.s347944] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 02/01/2023] [Indexed: 02/10/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignancy that requires multidisciplinary evaluation to develop individualized and tailored treatment concepts. While liver resection and transplantation represent the mainstay of curative treatment in patients with early-stage HCC, disease recurrence remains an important burden. Immune checkpoint inhibitors (ICI) have become standard of care in the palliative setting, achieving promising response rates with overall good tolerability. Accordingly, ICIs are being evaluated in (neo)adjuvant concepts in order to improve survival. Nevertheless, neoadjuvant therapies are not recommended by current guidelines as they have not been proven to improve the outcome in large Phase III trials yet. Especially in the context of liver transplantation (LT), perioperative ICI usage is in need of a particularly critical risk-benefit assessment, as the immunotherapy may significantly increase the risk of rejection. In this review, we summarize available data on ICI-based perioperative treatment strategies in HCC. We discuss current drawbacks and challenges of this treatment concept and specifically highlight the risk of allograft rejection when ICI are given in patients (subsequently) considered for liver transplantation.
Collapse
Affiliation(s)
- Alena Laschtowitz
- Department of Hepatology and Gastroenterology, Charité – Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | - Christoph Roderburg
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité – Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | - Raphael Mohr
- Department of Hepatology and Gastroenterology, Charité – Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany,Correspondence: Raphael Mohr, Department of Hepatology and Gastroenterology, Charité – Universitätsmedizin Berlin, Augustenburger Platz 1, Berlin, 13353, Germany, Email
| |
Collapse
|
33
|
Morganti S, Tolaney SM. Role of Immunotherapy in Early- and Late-Stage Triple-Negative Breast Cancer. Hematol Oncol Clin North Am 2023; 37:133-150. [PMID: 36435606 DOI: 10.1016/j.hoc.2022.08.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
For women with triple-negative breast cancer, the addition of pembrolizumab to chemotherapy has become a standard of care in the early-stage and first-line metastatic setting. However, many questions persist. Different chemotherapy backbones and sequencing strategies have been evaluated, but evidence supporting the superiority of one over the other is weak. Although many have been investigated, programmed cell death ligand 1 (PDL1) is the only approved biomarker. Since immunotherapy has been associated with potentially severe and permanent toxicities, the identification of better predictive biomarkers is essential. New strategies are needed to increase the proportion of patients who might benefit from immunotherapy.
Collapse
Affiliation(s)
- Stefania Morganti
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Boston, MA, USA; Department of Oncology and Hemato-Oncology, University of Milan, Istituto Europeo di Oncologia, Milan, Italy
| | - Sara M Tolaney
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
34
|
Loizides S, Constantinidou A. Triple negative breast cancer: Immunogenicity, tumor microenvironment, and immunotherapy. Front Genet 2023; 13:1095839. [PMID: 36712858 PMCID: PMC9879323 DOI: 10.3389/fgene.2022.1095839] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 12/30/2022] [Indexed: 01/15/2023] Open
Abstract
Triple negative breast cancer (TNBC) is a biologically diverse subtype of breast cancer characterized by genomic and transcriptional heterogeneity and exhibiting aggressive clinical behaviour and poor prognosis. In recent years, emphasis has been placed on the identification of mechanisms underlying the complex genomic and biological profile of TNBC, aiming to tailor treatment strategies. High immunogenicity, specific immune activation signatures, higher expression of immunosuppressive genes and higher levels of stromal Tumor Infiltrating Lymphocytes, constitute some of the key elements of the immune driven landscape associated with TNBC. The unprecedented response of TNBC to immunotherapy has undoubtedly changed the standard of care in this disease both in the early and the metastatic setting. However, the extent of interplay between immune infiltration and mutational signatures in TNBC is yet to be fully unravelled. In the present review, we present clinical evidence on the immunogenicity and tumour microenvironment influence on TNBC progression and the current treatment paradigms in TNBC based on immunotherapy.
Collapse
Affiliation(s)
- Sotiris Loizides
- Medical Oncology Department, Bank of Cyprus Oncology Centre, Nicosia, Cyprus
| | - Anastasia Constantinidou
- Medical Oncology Department, Bank of Cyprus Oncology Centre, Nicosia, Cyprus
- Medical School, University of Cyprus, Nicosia, Cyprus
- Cyprus Cancer Research Institute, Nicosia, Cyprus
| |
Collapse
|
35
|
Schreier A, Zappasodi R, Serganova I, Brown KA, Demaria S, Andreopoulou E. Facts and Perspectives: Implications of tumor glycolysis on immunotherapy response in triple negative breast cancer. Front Oncol 2023; 12:1061789. [PMID: 36703796 PMCID: PMC9872136 DOI: 10.3389/fonc.2022.1061789] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 11/17/2022] [Indexed: 01/11/2023] Open
Abstract
Triple negative breast cancer (TNBC) is an aggressive disease that is difficult to treat and portends a poor prognosis in many patients. Recent efforts to implement immune checkpoint inhibitors into the treatment landscape of TNBC have led to improved outcomes in a subset of patients both in the early stage and metastatic settings. However, a large portion of patients with TNBC remain resistant to immune checkpoint inhibitors and have limited treatment options beyond cytotoxic chemotherapy. The interplay between the anti-tumor immune response and tumor metabolism contributes to immunotherapy response in the preclinical setting, and likely in the clinical setting as well. Specifically, tumor glycolysis and lactate production influence the tumor immune microenvironment through creation of metabolic competition with infiltrating immune cells, which impacts response to immune checkpoint blockade. In this review, we will focus on how glucose metabolism within TNBC tumors influences the response to immune checkpoint blockade and potential ways of harnessing this information to improve clinical outcomes.
Collapse
Affiliation(s)
- Ashley Schreier
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY, United States
| | - Roberta Zappasodi
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, United States,Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, United States,Parker Institute for Cancer Immunotherapy, San Francisco, CA, United States
| | - Inna Serganova
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, United States,Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Kristy A. Brown
- Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Sandra Demaria
- Department of Radiation Oncology and Department of Pathology, Weill Cornell Medicine, New York, NY, United States
| | - Eleni Andreopoulou
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY, United States,*Correspondence: Eleni Andreopoulou,
| |
Collapse
|
36
|
Fasching PA, Hein A, Kolberg HC, Häberle L, Uhrig S, Rübner M, Belleville E, Hack CC, Fehm TN, Janni W, Hartmann A, Erber R, Theuser AK, Brucker SY, Hartkopf AD, Untch M. Pembrolizumab in combination with nab-paclitaxel for the treatment of patients with early-stage triple-negative breast cancer - A single-arm phase II trial (NeoImmunoboost, AGO-B-041). Eur J Cancer 2023; 184:1-9. [PMID: 36871424 DOI: 10.1016/j.ejca.2023.01.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 12/23/2022] [Accepted: 01/01/2023] [Indexed: 01/11/2023]
Abstract
BACKGROUND Pembrolizumab is approved for the neoadjuvant/adjuvant treatment of early triple-negative breast cancer (TNBC) patients in combination with chemotherapy. The Keynote-522 trial used platinum chemotherapy. As neoadjuvant nab-paclitaxel (nP) is also highly effective in triple-negative breast cancer patients, this study investigates the response to nP-containing neoadjuvant chemotherapy in combination with pembrolizumab. PATIENTS AND METHODS NeoImmunoboost (AGO-B-041/NCT03289819) is a multicenter, prospective single-arm phase II trial. Patients were treated with 12 weekly cycles of nP followed by four three-weekly cycles of epirubicin/cyclophosphamide. Pembrolizumab was given three-weekly in combination with these chemotherapies. The study was planned for 50 patients. After 25 patients, the study was amended to include a pre-chemotherapy single application of pembrolizumab. The primary aim was pathological complete response (pCR), and the secondary aims were safety and quality of life. RESULTS Of 50 included patients, 33 (66.0%; 95%confidence interval: 51.2%-78.8%) had a (ypT0/is ypN0) pCR. The pCR rate in the per-protocol population (n = 39) was 71.8% (95%confidence interval: 55.1%-85.0%). The most common adverse events of any grade were fatigue (58.5%), peripheral sensory neuropathy (54.7%) and neutropenia (52.8%). The pCR rate in the cohort of 27 patients with a pre-chemotherapy pembrolizumab dose was 59.3%, and 73.9% in the 23 patients without pre-chemotherapy dose. CONCLUSIONS pCR rates after NACT with nP and anthracycline combined with pembrolizumab are encouraging. With acceptable side-effect profiles, this treatment might be a reasonable alternative to platinum-containing chemotherapy in cases of contraindications. However, without data from randomised trials and long-term follow up, platinum/anthracycline/taxane-based chemotherapy remains the standard combination chemotherapy for pembrolizumab.
Collapse
Affiliation(s)
- Peter A Fasching
- Erlangen University Hospital, Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Germany.
| | - Alexander Hein
- Erlangen University Hospital, Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Germany
| | | | - Lothar Häberle
- Erlangen University Hospital, Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Germany; Biostatistics Unit, Department of Gynecology and Obstetrics, Erlangen University Hospital, Erlangen, Germany
| | - Sabrina Uhrig
- Erlangen University Hospital, Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Germany
| | - Matthias Rübner
- Erlangen University Hospital, Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Germany
| | | | - Carolin C Hack
- Erlangen University Hospital, Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Germany
| | - Tanja N Fehm
- Department of Gynecology and Obstetrics, University Hospital Düsseldorf, Germany
| | - Wolfang Janni
- Department of Gynecology and Obstetrics, Ulm University Hospital, Ulm, Germany
| | - Arndt Hartmann
- Erlangen University Hospital, Institute of Pathology, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Germany
| | - Ramona Erber
- Erlangen University Hospital, Institute of Pathology, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Germany
| | | | - Sara Y Brucker
- Department of Obstetrics and Gynecology, University of Tuebingen, Tuebingen, Germany
| | - Andreas D Hartkopf
- Department of Gynecology and Obstetrics, Ulm University Hospital, Ulm, Germany
| | - Michael Untch
- Clinic for Gynecology and Obstetrics, Breast Cancer Center, Gynecologic Oncology Center, Helios Klinikum Berlin Buch, Germany
| |
Collapse
|
37
|
Cancer Vaccines for Triple-Negative Breast Cancer: A Systematic Review. Vaccines (Basel) 2023; 11:vaccines11010146. [PMID: 36679991 PMCID: PMC9866612 DOI: 10.3390/vaccines11010146] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 01/12/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is the subtype of breast cancer with the poorest outcomes, and is associated with a high risk of relapse and metastasis. The treatment choices for this malignancy have been confined to conventional chemotherapeutic agents, due to a lack of expression of the canonical molecular targets. Immunotherapy has been recently changing the treatment paradigm for many types of tumors, and the approach of evoking active immune responses in the milieu of breast tumors through cancer vaccines has been introduced as one of the most novel immunotherapeutic approaches. Accordingly, a number of vaccines for the treatment or prevention of recurrence have been developed and are currently being studied in TNBC patients, while none have yet received any approvals. To elucidate the efficacy and safety of these vaccines, we performed a systematic review of the available literature on the topic. After searching the PubMed, Scopus, Web of Science, Embase, Cochrane CENTRAL, and Google Scholar databases, a total of 5701 results were obtained, from which 42 clinical studies were eventually included based on the predefined criteria. The overall quality of the included studies was acceptable. However, due to a lack of reporting outcomes of survival or progression in some studies (which were presented as conference abstracts) as well as the heterogeneity of the reported outcomes and study designs, we were not able to carry out a meta-analysis. A total of 32 different vaccines have so far been evaluated in TNBC patients, with the majority belonging to the peptide-based vaccine type. The other vaccines were in the cell or nucleic acid (RNA/DNA)-based categories. Most vaccines proved to be safe with low-grade, local adverse events and could efficiently evoke cellular immune responses; however, most trials were not able to demonstrate significant improvements in clinical indices of efficacy. This is in part due to the limited number of randomized studies, as well as the limited TNBC population of each trial. However, due to the encouraging results of the currently published trials, we anticipate that this strategy could show its potential through larger, phase III randomized studies in the near future.
Collapse
|
38
|
Immune-Related Gene Signatures to Predict the Effectiveness of Chemoimmunotherapy in Triple-Negative Breast Cancer Using Exploratory Subgroup Discovery. Cancers (Basel) 2022; 14:cancers14235806. [PMID: 36497286 PMCID: PMC9735620 DOI: 10.3390/cancers14235806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 11/23/2022] [Accepted: 11/23/2022] [Indexed: 11/26/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer with limited therapeutic options. Although immunotherapy has shown potential in TNBC patients, clinical studies have only demonstrated a modest response. Therefore, the exploration of immunotherapy in combination with chemotherapy is warranted. In this project we identified immune-related gene signatures for TNBC patients that may explain differences in patients' outcomes after anti-PD-L1+chemotherapy treatment. First, we ran the exploratory subgroup discovery algorithm on the TNBC dataset comprised of 422 patients across 24 studies. Secondly, we narrowed down the search to twelve homogenous subgroups based on tumor mutational burden (TMB, low or high), relapse status (disease-free or recurred), tumor cellularity (high, low and moderate), menopausal status (pre- or post) and tumor stage (I, II and III). For each subgroup we identified a union of the top 10% of genotypic patterns. Furthermore, we employed a multinomial regression model to predict significant genotypic patterns that would be linked to partial remission after anti-PD-L1+chemotherapy treatment. Finally, we uncovered distinct immune cell populations (T-cells, B-cells, Myeloid, NK-cells) for TNBC patients with various treatment outcomes. CD4-Tn-LEF1 and CD4-CXCL13 T-cells were linked to partial remission on anti-PD-L1+chemotherapy treatment. Our informatics pipeline may help to select better responders to chemoimmunotherapy, as well as pinpoint the underlying mechanisms of drug resistance in TNBC patients at single-cell resolution.
Collapse
|
39
|
Abdou Y, Goudarzi A, Yu JX, Upadhaya S, Vincent B, Carey LA. Immunotherapy in triple negative breast cancer: beyond checkpoint inhibitors. NPJ Breast Cancer 2022; 8:121. [PMID: 36351947 PMCID: PMC9646259 DOI: 10.1038/s41523-022-00486-y] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 10/13/2022] [Indexed: 11/10/2022] Open
Abstract
The development of immunotherapy agents has revolutionized the field of oncology. The only FDA-approved immunotherapeutic approach in breast cancer consists of immune checkpoint inhibitors, yet several novel immune-modulatory strategies are being actively studied and appear promising. Innovative immunotherapeutic strategies are urgently needed in triple negative breast cancer (TNBC), a subtype of breast cancer known for its poor prognosis and its resistance to conventional treatments. TNBC is more primed to respond to immunotherapy given the presence of more tumor infiltrating lymphocytes, higher PD-L1 expression, and higher tumor mutation burden relative to the other breast cancer subtypes, and therefore, immuno-oncology represents a key area of promise for TNBC research. The aim of this review is to highlight current data and ongoing efforts to establish the safety and efficacy of immunotherapeutic approaches beyond checkpoint inhibitors in TNBC.
Collapse
Affiliation(s)
- Yara Abdou
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA.
| | - Atta Goudarzi
- Department of Medicine, University at Buffalo, Buffalo, NY, 14203, USA
| | - Jia Xin Yu
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, 94129, USA
| | | | - Benjamin Vincent
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Lisa A Carey
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| |
Collapse
|
40
|
Agostinetto E, Jacobs F, Debien V, De Caluwé A, Pop CF, Catteau X, Aftimos P, de Azambuja E, Buisseret L. Post-Neoadjuvant Treatment Strategies for Patients with Early Breast Cancer. Cancers (Basel) 2022; 14:cancers14215467. [PMID: 36358886 PMCID: PMC9654353 DOI: 10.3390/cancers14215467] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/14/2022] [Accepted: 10/31/2022] [Indexed: 11/09/2022] Open
Abstract
Simple Summary Treatment strategies for early breast cancer have significantly improved in the last decades. Several new effective agents have proved clinical benefit and have entered the clinics, changing the treatment landscape for this disease and inducing significant prolongation of patient survival. Alongside, there has been an evolution in the design of clinical trials for early breast cancer, with an increasing interest in the pre-surgical treatment approach, which allows a direct evaluation of treatment effect on tumor size and a post-therapy risk stratification. Consequently, the post-neoadjuvant setting has been gaining increasing attention, thanks to the possibility to provide additional treatment for selected patients at higher risk of relapse, namely those who did not respond to neoadjuvant therapy and had residual disease at surgery. Abstract Pre-surgical treatments in patients with early breast cancer allows a direct estimation of treatment efficacy, by comparing the tumor and the treatment. Patients who achieve a pathological complete response at surgery have a better prognosis, with lower risk of disease recurrence and death. Hence, clinical research efforts have been focusing on high-risk patients with residual disease at surgery, who may be “salvaged” through additional treatments administered in the post-neoadjuvant setting. In the present review, we aim to illustrate the development and advantages of the post-neoadjuvant setting, and to discuss the available strategies for patients with early breast cancer, either approved or under investigation. This review was written after literature search on main scientific databases (e.g., PubMed) and conference proceedings from major oncology conferences up to 1 August 2022. T-DM1 and capecitabine are currently approved as post-neoadjuvant treatments for patients with HER2-positive and triple-negative breast cancer, respectively, with residual disease at surgery. More recently, other treatment strategies have been approved for patients with high-risk early breast cancer, including the immune checkpoint inhibitor pembrolizumab, the PARP inhibitor olaparib and the CDK 4/6 inhibitor abemaciclib. Novel agents and treatment combinations are currently under investigation as promising post-neoadjuvant treatment strategies.
Collapse
Affiliation(s)
- Elisa Agostinetto
- Institut Jules Bordet, L’Université Libre de Bruxelles (U.L.B.),1070 Bruxelles, Belgium
| | - Flavia Jacobs
- Institut Jules Bordet, L’Université Libre de Bruxelles (U.L.B.),1070 Bruxelles, Belgium
| | - Véronique Debien
- Institut Jules Bordet, L’Université Libre de Bruxelles (U.L.B.),1070 Bruxelles, Belgium
| | - Alex De Caluwé
- Institut Jules Bordet, L’Université Libre de Bruxelles (U.L.B.),1070 Bruxelles, Belgium
| | - Catalin-Florin Pop
- Institut Jules Bordet, L’Université Libre de Bruxelles (U.L.B.),1070 Bruxelles, Belgium
| | - Xavier Catteau
- Curepath Laboratory (CHU Tivoli, CHIREC), Rue de Borfilet 12A, 6040 Jumet, Belgium
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, route de Lennik 808, 1070 Brussels, Belgium
| | - Philippe Aftimos
- Institut Jules Bordet, L’Université Libre de Bruxelles (U.L.B.),1070 Bruxelles, Belgium
| | - Evandro de Azambuja
- Institut Jules Bordet, L’Université Libre de Bruxelles (U.L.B.),1070 Bruxelles, Belgium
| | - Laurence Buisseret
- Institut Jules Bordet, L’Université Libre de Bruxelles (U.L.B.),1070 Bruxelles, Belgium
- Correspondence:
| |
Collapse
|
41
|
Toxicity and Timing of Breast Radiation Therapy With Overlapping Systemic Therapies. Int J Radiat Oncol Biol Phys 2022; 114:377-381. [PMID: 36152640 DOI: 10.1016/j.ijrobp.2022.05.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 05/18/2022] [Indexed: 11/22/2022]
|
42
|
Trapani D, Ferraro E, Giugliano F, Boscolo Bielo L, Curigliano G, Burstein HJ. Postneoadjuvant treatment for triple-negative breast cancer. Curr Opin Oncol 2022; 34:623-634. [PMID: 35993306 DOI: 10.1097/cco.0000000000000893] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Triple-negative breast cancer (TNBC) has been conventionally associated with poor prognosis, as a result of limited therapeutic options. In the early setting, prognosis is informed by clinical-pathological factors; for patients receiving neoadjuvant treatments, pathological complete response (pCR) is the strongest factor. In this review, we mapped the landscape of clinical trials in the postneoadjuvant space, and identified three patterns of clinical trial design. RECENT FINDINGS For patients at higher risk, effective postneoadjuvant treatments are of paramount importance to address a high clinical need. Postneoadjuvant risk-adapted treatments have demonstrated to improve survival in patients at high of recurrence. SUMMARY Patients at high risk have indication for adjuvant treatment intensification, informed by baseline clinical, pathological or molecular factors (type 1 approach), on the presence, extent and molecular characteristics of the residual disease at the time of surgery (type 2) or on risk factors assessed in the postsurgical setting (type 3), for example, circulating tumour DNA. Most of the past trials were based on type 2 approaches, for example, with capecitabine and Olaparib. Few trials were based on a type 1 approach, notably pembrolizumab for early TNBC. The clinical validity of type 3 approaches is under investigation in several ongoing trials.
Collapse
Affiliation(s)
- Dario Trapani
- Department of Medical Oncology, Dana-Farber Cancer Institute
- Harvard Medical School, Boston, Massachusetts
| | - Emanuela Ferraro
- Breast Medicine Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Federica Giugliano
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Luca Boscolo Bielo
- Division of 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 Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Harold J Burstein
- Department of Medical Oncology, Dana-Farber Cancer Institute
- Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
43
|
Paluch-Shimon S, Cardoso F, Partridge AH, Abulkhair O, Azim HA, Bianchi-Micheli G, Cardoso MJ, Curigliano G, Gelmon KA, Gentilini O, Harbeck N, Kaufman B, Kim SB, Liu Q, Merschdorf J, Poortmans P, Pruneri G, Senkus E, Sirohi B, Spanic T, Sulosaari V, Peccatori F, Pagani O. ESO-ESMO fifth international consensus guidelines for breast cancer in young women (BCY5). Ann Oncol 2022; 33:1097-1118. [PMID: 35934170 DOI: 10.1016/j.annonc.2022.07.007] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 07/14/2022] [Accepted: 07/17/2022] [Indexed: 12/31/2022] Open
Abstract
We dedicate this manuscript in memory of a dear friend and colleague Bella Kaufman. The fifth International Consensus Symposium for Breast Cancer in Young Women (BCY5) took place virtually in October 2020, organized by the European School of Oncology (ESO) and the European Society of Medical Oncology (ESMO). Consensus recommendations for the management of breast cancer in young women were updated from BCY4 with incorporation of new evidence to inform the guidelines. Areas of research priorities as well as specificities in different geographic and minority populations were identified. This manuscript summarizes the ESO-ESMO international consensus recommendations, which are also endorsed by the European Society of Breast Specialists (EUSOMA).
Collapse
Affiliation(s)
- S Paluch-Shimon
- Hadassah University Hospital & Faculty of Medicine, Hebrew University, Jerusalem, Israel.
| | - F Cardoso
- Breast Unit, Champalimaud Clinical Centre/Champalimaud Foundation, Lisbon, Portugal
| | | | - O Abulkhair
- King Abdulaziz Medical City for National Guard, Riyadh, Saudi Arabia
| | - H A Azim
- Breast Cancer Center, Hospital Zambrano Hellion, Tecnologico de Monterrey, San Pedro Garza Garcia, Nuevo Leon, Mexico
| | | | - M J Cardoso
- Breast Unit, Champalimaud Clinical Centre/Champalimaud Foundation, Lisbon, Portugal
| | - G Curigliano
- European Institute of Oncology IRCCS, Milan; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - K A Gelmon
- British Columbia Cancer, Vancouver, Canada
| | | | - N Harbeck
- Breast Center, Department of OB&GYN and CCCMunich, LMU University Hospital, Munich, Germany
| | - B Kaufman
- Sheba Medical Center, Ramat Gan, Israel
| | - S B Kim
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Q Liu
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | | | - P Poortmans
- Iridium Netwerk, Department of Radiation Oncology & University of Antwerp, Faculty of Medicine and Health Sciences, Wilrijk-Antwerp, Belgium
| | - G Pruneri
- National Cancer Institute, IRCCS Foundation, Milan, Italy
| | - E Senkus
- Medical University of Gdansk, Gdansk, Poland
| | - B Sirohi
- Max Institute of Cancer Care, New Delhi and Gurgaon, India
| | - T Spanic
- Europa Donna Slovenia, Ljubljana, Slovenia
| | - V Sulosaari
- European Oncology Nursing Society (EONS) and Turku University of Applied Sciences, Turku, Finland
| | - F Peccatori
- European Institute of Oncology IRCCS, Milan; European Institute of Oncology IRCCS & European School of Oncology, Milan, Italy
| | - O Pagani
- Interdisciplinary Cancer Service Hospital Riviera-Chablais Rennaz, Vaud, Geneva University Hospitals, Lugano University, Swiss Group for Clinical Cancer Research (SAKK), Lugano, Switzerland
| |
Collapse
|
44
|
Walsh EM, Gucalp A, Patil S, Edelweiss M, Ross DS, Razavi P, Modi S, Iyengar NM, Sanford R, Troso-Sandoval T, Gorsky M, Bromberg J, Drullinsky P, Lake D, Wong S, DeFusco PA, Lamparella N, Gupta R, Tabassum T, Boyle LA, Arumov A, Traina TA. Adjuvant enzalutamide for the treatment of early-stage androgen-receptor positive, triple-negative breast cancer: a feasibility study. Breast Cancer Res Treat 2022; 195:341-351. [PMID: 35986801 PMCID: PMC10506398 DOI: 10.1007/s10549-022-06669-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 06/29/2022] [Indexed: 11/02/2022]
Abstract
PURPOSE Chemotherapy with or without immunotherapy remains the mainstay of treatment for triple-negative breast cancer (TNBC). A subset of TNBCs express the androgen receptor (AR), representing a potential new therapeutic target. This study assessed the feasibility of adjuvant enzalutamide, an AR antagonist, in early-stage, AR-positive (AR +) TNBC. METHODS This study was a single-arm, open-label, multicenter trial in which patients with stage I-III, AR ≥ 1% TNBC who had completed standard-of-care therapy were treated with enzalutamide 160 mg/day orally for 1 year. The primary objective of this study was to evaluate the feasibility of 1 year of adjuvant enzalutamide, defined as the treatment discontinuation rate of enzalutamide due to toxicity, withdrawal of consent, or other events related to tolerability. Secondary endpoints included disease-free survival (DFS), overall survival (OS), safety, and genomic features of recurrent tumors. RESULTS Fifty patients were enrolled in this study. Thirty-five patients completed 1 year of therapy, thereby meeting the prespecified trial endpoint for feasibility. Thirty-two patients elected to continue with an optional second year of treatment. Grade ≥ 3 treatment-related adverse events were uncommon. The 1-year, 2-year, and 3-year DFS were 94%, 92% , and 80%, respectively. Median OS has not been reached. CONCLUSION This clinical trial demonstrates that adjuvant enzalutamide is a feasible and well-tolerated regimen in patients with an early-stage AR + TNBC. Randomized trials in the metastatic setting may inform patient selection through biomarker development; longer follow-up is needed to determine the effect of anti-androgens on DFS and OS in this patient population.
Collapse
Affiliation(s)
- Elaine M Walsh
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA.
| | - Ayca Gucalp
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA
| | - Sujata Patil
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marcia Edelweiss
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dara S Ross
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pedram Razavi
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA
| | - Shanu Modi
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA
| | - Neil M Iyengar
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA
| | - Rachel Sanford
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA
| | - Tiffany Troso-Sandoval
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA
| | - Mila Gorsky
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA
| | - Jacqueline Bromberg
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA
| | - Pamela Drullinsky
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA
| | - Diana Lake
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA
| | - Serena Wong
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA
| | | | | | - Ranja Gupta
- Lehigh Valley Health Network Cancer Institute, Allentown, PA, USA
| | - Tasmila Tabassum
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA
| | - Leigh Ann Boyle
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA
| | - Artavazd Arumov
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA
| | - Tiffany A Traina
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, 300 East 66thStreet, New York, NY, USA
| |
Collapse
|
45
|
Al-Share B, Assad H, Abrams J, Deol A, Alavi A, Modi D, Kin A, Ratanatharathorn V, Uberti J, Ayash L. Role of High-Dose Adjuvant Chemotherapy Followed by Autologous Stem Cell Transplantation in Locally Advanced Triple-Negative Breast Cancer: A Retrospective Chart Review. JOURNAL OF ONCOLOGY 2022; 2022:3472324. [PMID: 36213836 PMCID: PMC9546640 DOI: 10.1155/2022/3472324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 06/13/2022] [Accepted: 06/16/2022] [Indexed: 11/17/2022]
Abstract
Purpose Women with locally advanced/high-risk triple-negative breast cancer treated with the current standard chemotherapy continue to have a poor prognosis. High-dose chemotherapy with autologous stem cell transplant as treatment for locally advanced/high-risk breast cancer remains controversial due to a lack of survival benefit seen in previous phase III trials. However, these trials evaluated a heterogeneous group of patients with different receptor subtypes. A marginal benefit was observed in certain subgroups. We report long-term outcomes of women with stage IIB or III triple-negative breast cancer treated with high-dose chemotherapy followed by autologous stem cell transplant at our institution between 1995 and 2001. Methods This is a retrospective analysis of stage IIB or stage III triple-negative breast cancer treated with high-dose chemotherapy followed by autologous stem cell transplant. We excluded women with hormone-positive, HER2/neu-positive/unknown, and/or metastatic disease prior to transplant as per updated AJCC 7th edition guidelines. Patients underwent surgery and either neoadjuvant or adjuvant anthracycline and taxane-based chemotherapy and then proceeded to high-dose chemotherapy and autologous stem cell transplant using carmustine 600 mg/sqm, cyclophosphamide 5.6gm/sqm, and cisplatin 165 mg/sqm (STAMP 1 regimen) for consolidation. This was followed by locoregional breast and lymph node radiation per standard of care. Results Twenty-nine women (2 stage IIB and 27 stage III) were evaluated. The median age at diagnosis was 43 years (IQR: 40, 51). Eleven patients had 4-9 regional lymph nodes (LN) involved and 16 had 10+ involved LNs. Four patients had T4 or inflammatory breast cancer and two had ipsilateral supraclavicular LNs involved. The median follow-up time is 16 years (95% CI: 12, 19, range <1-19 y) posttransplant. The median overall survival was 15 years (95% CI: 3, 19); the median DFS was 14 years (95% CI: 1, 19). Conclusions This study of locally advanced/high-risk triple-negative breast cancer treated with adjuvant high-dose chemotherapy and autologous stem cell transplant reveals high overall survival rate. With the current improvement in treatment-related mortality, re-evaluating this approach in this subset of high-risk breast cancer in prospective randomized studies may be worthwhile.
Collapse
Affiliation(s)
- Bayan Al-Share
- Department of Oncology, Wayne State University/Karmanos Cancer Institute, Detroit, MI, USA
| | - Hadeel Assad
- Department of Oncology, Wayne State University/Karmanos Cancer Institute, Detroit, MI, USA
| | - Judith Abrams
- Department of Oncology, Wayne State University/Karmanos Cancer Institute, Detroit, MI, USA
| | - Abhinav Deol
- Department of Oncology, Wayne State University/Karmanos Cancer Institute, Detroit, MI, USA
| | - Asif Alavi
- Department of Oncology, Wayne State University/Karmanos Cancer Institute, Detroit, MI, USA
| | - Dipenkumar Modi
- Department of Oncology, Wayne State University/Karmanos Cancer Institute, Detroit, MI, USA
| | - Andrew Kin
- Department of Oncology, Wayne State University/Karmanos Cancer Institute, Detroit, MI, USA
| | | | - Joseph Uberti
- Department of Oncology, Wayne State University/Karmanos Cancer Institute, Detroit, MI, USA
| | - Lois Ayash
- Department of Oncology, Wayne State University/Karmanos Cancer Institute, Detroit, MI, USA
| |
Collapse
|
46
|
Fehm TN, Welslau M, Müller V, Lüftner D, Schütz F, Fasching PA, Janni W, Thomssen C, Witzel I, Belleville E, Untch M, Thill M, Tesch H, Ditsch N, Lux MP, Aktas B, Banys-Paluchowski M, Schneeweiss A, Kolberg-Liedtke C, Hartkopf AD, Wöckel A, Kolberg HC, Harbeck N, Stickeler E. Update Breast Cancer 2022 Part 3 - Early-Stage Breast Cancer. Geburtshilfe Frauenheilkd 2022; 82:912-921. [PMID: 36110894 PMCID: PMC9470293 DOI: 10.1055/a-1912-7105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 07/31/2022] [Indexed: 11/01/2022] Open
Abstract
This review summarizes recent developments in the prevention and treatment of patients with early-stage breast cancer. The individual disease risk for different molecular subtypes was investigated in a large epidemiological study. With regard to treatment, new data are available from long-term follow-up of the Aphinity study, as well as new data on neoadjuvant therapy with atezolizumab in HER2-positive patients. Biomarkers, such as residual cancer burden, were investigated in the context of pembrolizumab therapy. A Genomic Grade Index study in elderly patients is one of a group of studies investigating the use of modern multigene tests to identify patients with an excellent prognosis in whom chemotherapy may be avoided. These and other aspects of the latest developments in the diagnosis and treatment of breast cancer are described in this review.
Collapse
Affiliation(s)
- Tanja N. Fehm
- Department of Gynecology and Obstetrics, University Hospital Düsseldorf, Düsseldorf, Germany
| | | | - Volkmar Müller
- Department of Gynecology, Hamburg-Eppendorf University Medical Center, Hamburg, Germany
| | - Diana Lüftner
- Immanuel Hospital Märkische Schweiz & Medical University of Brandenburg Theodor-Fontane, Brandenburg, Buckow, Germany
| | - Florian Schütz
- Gynäkologie und Geburtshilfe, Diakonissen-Stiftungs-Krankenhaus Speyer, Speyer, Germany
| | - Peter A. Fasching
- Erlangen University Hospital, Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen,
Germany,Correspondence/Korrespondenzadresse Peter A. Fasching, MD Erlangen University Hospital, Department of Gynecology and ObstetricsComprehensive Cancer
Center Erlangen EMNFriedrich Alexander University of Erlangen-NurembergUniversitätsstraße 21 – 2391054
ErlangenGermany
| | - Wolfgang Janni
- Department of Gynecology and Obstetrics, Ulm University Hospital, Ulm, Germany
| | - Christoph Thomssen
- Department of Gynaecology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Isabell Witzel
- Department of Gynecology, Hamburg-Eppendorf University Medical Center, Hamburg, Germany
| | | | - Michael Untch
- Clinic for Gynecology and Obstetrics, Breast Cancer Center, Gynecologic Oncology Center, Helios Klinikum Berlin Buch, Berlin, Germany
| | - Marc Thill
- Agaplesion Markus Krankenhaus, Department of Gynecology and Gynecological Oncology, Frankfurt am Main, Germany
| | - Hans Tesch
- Oncology Practice at Bethanien Hospital, Frankfurt am Main, Germany
| | - Nina Ditsch
- Department of Gynecology and Obstetrics, University Hospital Augsburg, Augsburg, Germany
| | - Michael P. Lux
- Klinik für Gynäkologie und Geburtshilfe, Frauenklinik St. Louise, Paderborn, St. Josefs-Krankenhaus, Salzkotten, St. Vincenz Krankenhaus GmbH, Germany
| | - Bahriye Aktas
- Department of Gynecology, University of Leipzig Medical Center, Leipzig, Germany
| | - Maggie Banys-Paluchowski
- Department of Gynecology and Obstetrics, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Andreas Schneeweiss
- National Center for Tumor Diseases (NCT), Heidelberg University Hospital and German Cancer Research Center, Heidelberg, Germany
| | | | - Andreas D. Hartkopf
- Department of Gynecology and Obstetrics, Ulm University Hospital, Ulm, Germany
| | - Achim Wöckel
- Department of Gynecology and Obstetrics, University Hospital Würzburg, Würzburg, Germany
| | | | - Nadia Harbeck
- Breast Center, Department of Gynecology and Obstetrics and CCC Munich LMU, LMU University Hospital, Munich, Germany
| | - Elmar Stickeler
- Department of Gynecology and Obstetrics, RWTH University Hospital Aachen, Aachen, Germany
| |
Collapse
|
47
|
Abstract
PURPOSE OF REVIEW Recently, immune checkpoint inhibitors (ICI) have demonstrated survival benefits in triple-negative breast cancer (TNBC) patients, treated in both the advanced and the early settings. RECENT FINDINGS As monotherapy, ICI failed to demonstrate a superiority over chemotherapy in pretreated advanced TNBC. In the first-line setting, ICI in combination with chemotherapy have shown consistent gains in progression-free survival in programmed death-ligand 1-positive TNBC, but only pembrolizumab indisputably demonstrated a significant overall survival benefit. In early-stage TNBC patients treated with neoadjuvant chemotherapy (NAC), ICI may improve the pathological complete response (pCR) rate. In the KEYNOTE-522 trial enrolling stage II to III TNBC patients, pembrolizumab, in combination with a NAC composed of carboplatin-paclitaxel followed by anthracyclines, and continued in the adjuvant phase led to significant increases in both pCR and disease-free survival, a practice-changing result in the field. Importantly, no unexpected safety signal was observed, but the possibility of definitive ICI-related toxicities may be challenging in curable early disease. SUMMARY Immunotherapy is now an important component in the therapeutic management of TNBC. Unresolved issues include the best chemotherapy partners, additional biomarkers to maximize the clinical benefit, and the possible extension of its use to other breast cancer subtypes.
Collapse
|
48
|
Schettini F, Venturini S, Giuliano M, Lambertini M, Pinato DJ, Elisa Onesti C, De Placido P, Harbeck N, Lüftner D, Denys H, Van Dam P, Arpino G, Zaman K, Mustacchi G, Gligorov J, Awada A, Campone M, Wildiers H, Gennari A, Tjan-Heijnen V, Bartsch R, Cortes J, Paris I, Martín M, De Placido S, Del Mastro L, Jerusalem G, Curigliano G, Prat A, Generali D. Multiple Bayesian Network Meta-Analyses to Establish Therapeutic Algorithms for Metastatic Triple Negative Breast Cancer. Cancer Treat Rev 2022; 111:102468. [DOI: 10.1016/j.ctrv.2022.102468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 09/20/2022] [Accepted: 09/22/2022] [Indexed: 12/23/2022]
|
49
|
Azim HA, Shohdy KS, Elghazawy H, Salib MM, Almeldin D, Kassem L. Programmed death-ligand 1 (PD-L1) expression predicts response to neoadjuvant chemotherapy in triple-negative breast cancer: A systematic review and meta-analysis. Biomarkers 2022; 27:764-772. [PMID: 35980714 DOI: 10.1080/1354750x.2022.2112614] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Background: In patients with metastatic triple-negative breast cancer (TNBC), PD-L1 expression has been demonstrated to predict response to immunotherapy. It is unclear whether PD-L1 expression measured with currently available validated assays can predict chemotherapy response in patients with non-metastatic TNBC.Methods: We conducted a systematic review and meta-analysis of clinical studies to assess the PD-L1 expression as a predictor of response to chemotherapy in non-metastatic TNBC using validated assays. The primary endpoint was pathological complete response (pCR) rate to neoadjuvant chemotherapy. Secondary endpoints included the prevalence of PD-L1 expression in non-metastatic TNBC and its impact on disease-free survival (DFS) and overall survival (OS). Moreover, RNA sequence data from the TCGA breast cancer cohort was used to define the relationship between PDCD1 and response to chemotherapy and prognosis.Results: Nineteen studies were eligible for the meta-analysis with a total of 2403 patients with non-metastatic TNBC disease. The PD-L1-positive cohort had a significantly higher likelihood of achieving pCR with neoadjuvant chemotherapy (pooled odds ratio =1.95; 95% CI= 1.39-2.73, p <0.0001). In studies which reported long-term outcomes, PD-L1 positivity was associated with significantly better DFS and OS compared to PD-L1 negative patients (pooled hazard ratio= 0.51; 95% CI= 0.35-0.74, p< 0.0001 and 0.51; 95% CI= 0.27-0.94, p = 0.031, respectively). Transcriptomic data suggested that PD-L1 expression is a surrogate marker for the upregulation of key immune-related genes that mediate response to chemotherapy in TNBC.Conclusion: This analysis clearly shows that patients with PD-L1 positive TNBC respond better to neoadjuvant chemotherapy and are associated with better survival outcomes compared to patients with PD-L1 negative tumors. The newly distinct quadruple negative breast cancer (QNBC) subtype should be defined as the BC subtype with the poorest outcome in the non-metastatic setting, highlighting the need for more aggressive therapy approaches.
Collapse
Affiliation(s)
- Hamdy A Azim
- Clinical Oncology Department, Cairo University, Cairo, Egypt
| | - Kyrillus S Shohdy
- Clinical Oncology Department, Cairo University, Cairo, Egypt.,Experimental Cancer Medicine Team, The Christie NHS Foundation Trust, Manchester, UK
| | - Hagar Elghazawy
- Clinical Oncology Department, Ain Shams University, Cairo, Egypt
| | - Monica M Salib
- Clinical Oncology Department, Cairo University, Cairo, Egypt
| | - Doaa Almeldin
- Clinical Oncology Department, Cairo University, Cairo, Egypt
| | - Loay Kassem
- Clinical Oncology Department, Cairo University, Cairo, Egypt
| |
Collapse
|
50
|
Howard FM, Pearson AT, Nanda R. Clinical trials of immunotherapy in triple-negative breast cancer. Breast Cancer Res Treat 2022; 195:1-15. [PMID: 35834065 PMCID: PMC9338129 DOI: 10.1007/s10549-022-06665-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 06/23/2022] [Indexed: 01/12/2023]
Abstract
PURPOSE Immunotherapy has started to transform the treatment of triple-negative breast cancer (TNBC), in part due to the unique immunogenicity of this breast cancer subtype. This review summarizes clinical studies of immunotherapy in advanced and early-stage TNBC. FINDINGS Initial studies of checkpoint blockade monotherapy demonstrated occasional responses, especially in patients with untreated programmed death-ligand 1 (PD-L1) positive advanced TNBC, but failed to confirm a survival advantage over chemotherapy. Nonetheless, pembrolizumab monotherapy has tumor agnostic approval for microsatellite instability-high or high tumor mutational burden cancers, and thus can be considered for select patients with advanced TNBC. Combination chemoimmunotherapy approaches have been more successful, and pembrolizumab is approved for PD-L1 positive advanced TNBC in combination with chemotherapy. This success has been translated to the curative setting, where pembrolizumab is now approved in combination with neoadjuvant chemotherapy for high-risk early-stage TNBC. CONCLUSION Immunotherapy has been a welcome addition to the growing armamentarium for TNBC, but responses remain limited to a subset of patients. Innovative strategies are under investigation in an attempt to induce immune responses in resistant tumors-with regimens incorporating small-molecule inhibitors, novel immune checkpoint targets, and intratumoral injections that directly alter the tumor microenvironment. As the focus shifts toward the use of immunotherapy for early-stage TNBC, it will be critical to identify those who derive the most benefit from treatment, given the potential for irreversible autoimmune toxicity and the lack of predictive accuracy of PD-L1 expression in the early-stage setting.
Collapse
Affiliation(s)
- Frederick M Howard
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medicine & Biological Sciences, 5841 S. Maryland Ave MC 2115, Chicago, IL, 60637, USA.
| | - Alexander T Pearson
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medicine & Biological Sciences, 5841 S. Maryland Ave MC 2115, Chicago, IL, 60637, USA
| | - Rita Nanda
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medicine & Biological Sciences, 5841 S. Maryland Ave MC 2115, Chicago, IL, 60637, USA
| |
Collapse
|