1
|
Wang Z, You P, Yang Z, Xiao H, Tang X, Pan Y, Li X, Gao F. PD-1/PD-L1 immune checkpoint inhibitors in the treatment of unresectable locally advanced or metastatic triple negative breast cancer: a meta-analysis on their efficacy and safety. BMC Cancer 2024; 24:1339. [PMID: 39478479 PMCID: PMC11526542 DOI: 10.1186/s12885-024-13105-9] [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: 10/05/2024] [Accepted: 10/24/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Triple negative breast cancer (TNBC) represents a particularly aggressive and clinically challenging subtype of breast cancer, characterized by its invasive nature and generally poor prognosis. Treatment options for unresectable TNBC are limited. In recent years, the advent of PD-1/PD-L1 immune checkpoint inhibitors has offered a promising new treatment option for unresectable TNBC. The role of PD-1/PD-L1 immune checkpoint inhibitors (ICIs) in unresectable TNBC management remains a subject of debate. This article aims to synthesize evidence from randomized controlled trials (RCTs) through a meta-analysis (MA) to provide a comprehensive evaluation of the efficacy and safety profile of ICIs in the treatment of unresectable TNBC. METHOD We searched PubMed, Embase, Cochrane library, Web of Science, and ClinicalTrials.gov for the eligible RCTs which compared the efficacy and safety of PD-1/PD-L1 ICIs and chemotherapy alone. The outcomes analyzed included overall survival (OS), progression-free survival (PFS), objective response rate (ORR) and treatment-related adverse effects (AEs). RESULTS This meta-analysis included 11 trials. Therapy with PD-L1 inhibitors was superior to chemotherapy in terms of OS in both the intention-to-treat (ITT) population and the PD-L1-positive population. (ITT: HR: = 0.90 [0.81, 0.99], P = 0.04, I2 = 48%; PD-L1 + : HR = 0.82 [0.70, 0.95], P = 0.01, I2 = 64%); In terms of PFS, treatment with PD-L1 inhibitors prolonged PFS in both the ITT and PD-L1-positive populations compared with chemotherapy (ITT: HR: = 0.85 [0.77, 0.93], P = 0.0006, I2 = 46%; PD-L1 + : HR = 0.72 [0.62, 0.83], P < 0.00001, I2 = 70%, Fig. 5); Compared with chemotherapy alone, treatment with PD-1 inhibitors prolonged OS in both the PD-L1-positive and ITT populations. (ITT: HR = 0.87 [0.78, 0.96], P = 0.007, I2 = 71%; CPS ≥ 1: HR = 0.81 [0.71, 0.92], P = 0.001, I2 = 39%); In terms of PFS, therapy with PD-1 inhibitors improved PFS in both the ITT population and the PD-L1-positive population compared with chemotherapy. (ITT: HR = 0.79 [0.70, 0.90], P = 0.0004, I2 = 0%; CPS ≥ 1: HR = 0.71 [0.61, 0.83], P < 0.0001, I2 = 0%; CPS ≥ 10: HR = 0.67 [0.53, 0.84], P = 0.0008, I2 = 0%). CONCLUSION Both PD-1 and PD-L1 inhibitors can offer survival benefits to TNBC patients, with the primary beneficiaries being those who are PD-L1-positive. However, immunotherapy can also lead to an increase in treatment-related adverse events. Therefore, it is essential to conduct a risk assessment for each patient before starting treatment to prevent the occurrence of serious adverse reactions. TRIAL REGISTRATION This systematic review study has been filed with PROSPERO (Registration number: CRD42024571775).
Collapse
Affiliation(s)
- Zuxiu Wang
- Department of General Practice Medicine, Second People's Hospital of Bengbu City, No. 302 Yan'an Road, Bengbu, Anhui Province, China
| | - Peimeng You
- Department of Biomedical Engineering, Capital Medical University, Beijing, China
| | - Zhanglei Yang
- Department of Rehabilitation Medicine, Red Cross Hospital Affiliated to Xi'an Jiaotong University, Xi'an, China
| | - Hanxi Xiao
- Department of General Practice Medicine, Nanchang University Affiliated Rehabilitation Hospital, Square South Road, Xihu District, Nanchang, Jiangxi Province, China
| | - Xinrong Tang
- Department of General Practice Medicine, Nanchang University Affiliated Rehabilitation Hospital, Square South Road, Xihu District, Nanchang, Jiangxi Province, China
| | - Yongping Pan
- Department of General Practice Medicine, Nanchang University Affiliated Rehabilitation Hospital, Square South Road, Xihu District, Nanchang, Jiangxi Province, China
| | - Xuhuan Li
- Department of General Practice Medicine, Nanchang University Affiliated Rehabilitation Hospital, Square South Road, Xihu District, Nanchang, Jiangxi Province, China.
| | - Feng Gao
- Department of General Practice Medicine, Second People's Hospital of Bengbu City, No. 302 Yan'an Road, Bengbu, Anhui Province, China.
| |
Collapse
|
2
|
Im SA, Cortes J, Cescon DW, Yusof MM, Iwata H, Masuda N, Takano T, Huang CS, Chung CF, Tsugawa K, Park YH, Matsumoto K, Inoue K, Kwong A, Loi S, Fu W, Pan W, Karantza V, Rugo HS, Schmid P. Results from the randomized KEYNOTE-355 study of pembrolizumab plus chemotherapy for Asian patients with advanced TNBC. NPJ Breast Cancer 2024; 10:79. [PMID: 39266535 PMCID: PMC11393332 DOI: 10.1038/s41523-024-00679-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 07/19/2024] [Indexed: 09/14/2024] Open
Abstract
In the phase 3 KEYNOTE-355 study (NCT02819518), pembrolizumab plus chemotherapy demonstrated statistically significant and clinically meaningful improvements in progression-free survival (PFS) and overall survival (OS) versus placebo plus chemotherapy among patients with previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (TNBC) and programmed cell death ligand 1 (PD-L1) combined positive score (CPS) ≥ 10 tumors. We analyzed outcomes for the subgroup of patients enrolled in Asia in KEYNOTE-355. Patients received pembrolizumab 200 mg or placebo (2:1 randomization) every 3 weeks for 35 cycles plus investigator's choice chemotherapy. Primary endpoints were PFS per Response Evaluation Criteria in Solid Tumors version 1.1 and OS. Among patients enrolled in Hong Kong, Japan, Korea, Malaysia and Taiwan (pembrolizumab plus chemotherapy, n = 113; placebo plus chemotherapy, n = 47), 117 (73.1%) had PD-L1 CPS ≥ 1 and 56 (35.0%) had PD-L1 CPS ≥ 10. Median time from randomization to data cutoff (June 15, 2021) was 43.8 (range, 36.8‒53.2) months (intent-to-treat [ITT] population). Hazard ratios (HRs [95% CI]) for PFS in the CPS ≥ 10, CPS ≥ 1, and ITT populations were 0.48 (0.24‒0.98), 0.58 (0.37‒0.91), and 0.66 (0.44‒0.99), respectively. Corresponding HRs (95% CI) for OS were 0.54 (0.28‒1.04), 0.62 (0.40‒0.97), and 0.57 (0.39‒0.84). Grade 3/4 treatment-related adverse events (AEs) occurred in 77.9% versus 78.7% of patients with pembrolizumab plus chemotherapy versus placebo plus chemotherapy. No grade 5 AEs occurred. Clinically meaningful improvement in PFS and OS with manageable toxicity were observed with pembrolizumab plus chemotherapy versus placebo plus chemotherapy in patients enrolled in Asia with previously untreated, inoperable or metastatic TNBC.Trial registration: ClinicalTrials.gov, NCT02819518.
Collapse
Affiliation(s)
- Seock-Ah Im
- Seoul National University Hospital, Seoul National University College of Medicine, Cancer Research Institute, Seoul National University, Seoul, Republic of Korea.
| | - Javier Cortes
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, Madrid and Barcelona, Spain
- Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid, Spain
| | - David W Cescon
- Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Mastura Md Yusof
- Cancer Center at Pantai Hospital Kuala Lumpur, Kuala Lumpur, Malaysia
| | - Hiroji Iwata
- Aichi Cancer Center Hospital, Nagoya, Aichi, Japan
| | - Norikazu Masuda
- Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Toshimi Takano
- The Cancer Institute Hospital of JFCR, Tokyo, Japan
- Toranomon Hospital, Tokyo, Japan
| | - Chiun-Sheng Huang
- National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chi-Feng Chung
- Koo Foundation Sun Yat-Sen Cancer Center, Taipei, Taiwan
| | | | - Yeon Hee Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | | | | | - Ava Kwong
- Division of Breast Surgery, The University of Hong Kong, Queen Mary and Tung Wah Hospital, Hong Kong, China
- The University of Hong Kong-ShenZhen Hospital, Shenzhen, China
| | - Sherene Loi
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Australia
- The Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Parkville, Australia
| | - Wei Fu
- Merck & Co., Inc., Rahway, NJ, USA
| | | | | | - Hope S Rugo
- University of California San Francisco Comprehensive Cancer Center, San Francisco, CA, USA
| | - Peter Schmid
- Centre for Experimental Cancer Medicine, Barts Cancer Institute, London, UK
| |
Collapse
|
3
|
Zhang L, Bai H, Zhou J, Ye L, Gao L. Role of tumor cell pyroptosis in anti-tumor immunotherapy. CELL INSIGHT 2024; 3:100153. [PMID: 38464416 PMCID: PMC10924176 DOI: 10.1016/j.cellin.2024.100153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 01/28/2024] [Accepted: 01/30/2024] [Indexed: 03/12/2024]
Abstract
Peripheral tumor-specific CD8+ T cells often fail to infiltrate into tumor parenchyma due to the immunosuppression of tumor microenvironment (TME). Meanwhile, a significant portion of tumor-specific CD8+ T cells infiltrated into TME are functionally exhausted. Despite the enormous success of anti-PD-1/PD-L1 immune-checkpoint blockade (ICB) treatment in a wide variety of cancer types, the majority of patients do not respond to this treatment largely due to the failure to efficiently drive tumor-specific CD8+ T cell infiltration and reverse their exhaustion states. Nowadays, tumor cell pyroptosis, a unique cell death executed by pore-forming gasdermin (GSDM) family proteins dependent or independent on inflammatory caspase activation, has been shown to robustly promote immune-killing of tumor cells by enhancing tumor immunogenicity and altering the inflammatory state in the TME, which would be beneficial in overcoming the shortages of anti-PD-1/PD-L1 ICB therapy. Therefore, in this review we summarize the current progresses of tumor cell pyroptosis in enhancing immune function and modulating TME, which synergizes anti-PD-1/PD-L1 ICB treatment to achieve better anti-tumor effect. We also enumerate several strategies to better amply the efficiency of anti-PD-1/PD-L1 ICB therapy by inducing tumor cell pyroptosis.
Collapse
Affiliation(s)
- Lincheng Zhang
- Institute of Immunology, Third Military Medical University, Chongqing, 400030, China
| | - Haotian Bai
- Division of Natural and Applied Sciences, Duke Kunshan University, 8 Duke Ave, Kunshan, 215316, China
| | - Jing Zhou
- Institute of Immunology, Third Military Medical University, Chongqing, 400030, China
| | - Lilin Ye
- Institute of Immunology, Third Military Medical University, Chongqing, 400030, China
| | - Leiqiong Gao
- Institute of Immunological Innovation and Translation, Chongqing Medical University, Chongqing, 400030, China
| |
Collapse
|
4
|
Battogtokh G, Obidiro O, Akala EO. Recent Developments in Combination Immunotherapy with Other Therapies and Nanoparticle-Based Therapy for Triple-Negative Breast Cancer (TNBC). Cancers (Basel) 2024; 16:2012. [PMID: 38893132 PMCID: PMC11171312 DOI: 10.3390/cancers16112012] [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: 04/23/2024] [Revised: 05/17/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Triple-negative breast cancer (TNBC), lacking specific receptors found in other breast cancer subtypes, poses significant treatment challenges due to limited therapeutic options. Therefore, it is necessary to develop novel treatment approaches for TNBC. In the last few decades, many attempts have been reported for alternative tools for TNBC treatment: immunotherapy, radiotherapy, targeted therapy, combination therapy, and nanotechnology-based therapy. Among them, combination therapy and nanotechnology-based therapy show the most promise for TNBC treatment. This review outlines recent advancements in these areas, highlighting the efficacy of combination therapy (immunotherapy paired with chemotherapy, targeted therapy, or radiotherapy) in both preclinical and clinical stages and nanotechnology-based therapies utilizing various nanoparticles loaded with anticancer agents, nucleic acids, immunotherapeutics, or CRISPRs in preclinical stages for TNBC treatment.
Collapse
Affiliation(s)
| | | | - Emmanuel O. Akala
- Center for Drug Research and Development, Department of Pharmaceutical Sciences, College of Pharmacy, Howard University, Washington, DC 20059, USA; (G.B.); (O.O.)
| |
Collapse
|
5
|
Lei C, Kong X, Li Y, Yang H, Zhang K, Wang Z, Chang H, Xuan L. PD-1/PD-L1 Inhibitor - Related Adverse Events and Their Management in Breast Cancer. J Cancer 2024; 15:2770-2787. [PMID: 38577606 PMCID: PMC10988294 DOI: 10.7150/jca.85433] [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: 04/19/2023] [Accepted: 03/03/2024] [Indexed: 04/06/2024] Open
Abstract
As the positive results of multiple clinical trials were released, the Programmed cell death 1 (PD-1) and Programmed cell death ligand 1 (PD-L1) inhibitors emerge as the focus of integrative breast cancer treatment. PD-1/PD-L1 inhibitors are often used as a sequential agent to be combined with other agents such as chemotherapeutic agents, targeted agents, and radiation therapy. As multiple therapies are administered simultaneously or in sequence, they are prone to a variety of adverse effects on patients while achieving efficacy. It is a challenge for clinicians to maintaining the balance between immune-related adverse effects(irAEs) and treatment efficacy. Previous literatures have paid lots of attention on the adverse effects caused by immunosuppressive agents themselves, while there is a dearth of the research on the management of adverse immune effects during the combination of immunotherapy with other treatments. In this review, we discuss the overall incidence of irAEs caused by PD-1/PD-L1 inhibitors in combination with various types of treatments in breast cancer, including chemotherapy, CTLA-4 inhibitors, targeted therapy, and radiotherapy, and systematically summarizes the clinical management to each organ-related adverse immune reaction. It is important to emphasize that in the event of irAEs such as neurological, hematologic, and cardiac toxicity, there is no alternative treatment but to terminate immunotherapy. Thus, seeking more effective strategy of irAEs' management is imminent and clinicians are urged to raise the awareness of the management of adverse immune reactions.
Collapse
Affiliation(s)
- Chuqi Lei
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiangyi Kong
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuan Li
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huaiyu Yang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ke Zhang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhongzhao Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hu Chang
- Administration Office, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lixue Xuan
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
6
|
Hosonaka K, Yamaoka K, Ikeda N, Uchida M, Uesawa Y, Takahashi K, Shimizu T. Disproportionality Analysis of Stomatitis Associated with Anticancer Drugs Using the Japanese Adverse Drug Event Report Database. Oncology 2024; 102:810-818. [PMID: 38198784 DOI: 10.1159/000535331] [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: 08/22/2023] [Accepted: 11/16/2023] [Indexed: 01/12/2024]
Abstract
INTRODUCTION Anticancer drug-induced stomatitis can affect a patient's quality of life and the continuation of drug treatment. Although there have been reports of the occurrence of stomatitis associated with anticancer agents in clinical trials, few Japanese participants have been enrolled in clinical trials and have not been sufficiently investigated. In addition, there has been little attention on research on anticancer drugs associated with stomatitis by patient stratification with different carcinogenic sites. Therefore, the aim of this study was to determine the disproportionality associated with stomatitis for various types of anticancer drugs in different types of cancer patients using the Japanese Adverse Drug Event Report (JADER) database. METHODS The aim of this study was to identify the disproportionality of stomatitis by analyzing the type of anticancer drug and cancer patients using the Japanese Pharmacovigilance Database. Data obtained from spontaneous reports of adverse events with more than 10 stomatitis outbreaks reported in the JADER database between April 2004 and March 2023 were analyzed. The safety signal for an adverse event was defined as the lower limit of the 95% confidence interval of the reported odds ratio of >1. RESULTS There were 6,178 reports of drugs associated with stomatitis. Among these, 41 drugs were suggested to be associated with stomatitis, and 41 drugs were detected as signals. These drugs were classified based on their efficacy: antipyrimidines (six drugs), folate metabolism antagonists (three drugs), alkylating agents (four drugs), platinum (three drugs), topoisomerase inhibitors (three drugs), microtubule inhibitors (three drugs), mammalian target of rapamycin (mTOR) inhibitors (two drugs), kinase inhibitors (seven drugs), anti-growth factor antibodies (five drugs) immune checkpoint inhibitors (one drug), and others (four drugs). CONCLUSION The drugs that may be associated with stomatitis were cell cycle-dependent drugs, epidermal growth factor receptor-tyrosine kinase inhibitors, and mTOR inhibitors. Moreover, this study suggested that anti-growth factor antibodies and immune checkpoint inhibitors may be associated with stomatitis development.
Collapse
Affiliation(s)
- Kousuke Hosonaka
- School of Pharmacy, Hyogo Medical University, Kobe, Japan
- Department of Pharmacy, Osaka-fu Saiseikai Noe Hospital, Osaka, Japan
| | - Kenta Yamaoka
- School of Pharmacy, Hyogo Medical University, Kobe, Japan
- Department of Pharmacy, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Naoe Ikeda
- School of Pharmacy, Hyogo Medical University, Kobe, Japan
| | - Mayako Uchida
- Department of Education and Research Center for Pharmacy Practice, Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, Kyotanabe, Japan
| | - Yoshihiro Uesawa
- Department of Medical Molecular Informatics, Meiji Pharmaceutical University, Kiyose, Japan
| | | | | |
Collapse
|
7
|
Hunter E, Salter M, Powell R, Dring A, Naithani T, Chatziioannou ME, Gebregzabhar A, Issa M, Green J, Ng S, Lim CR, Keat CS, Suan AT, Raman R, Fatt HK, Luen FLW, Alshaker H, Pchejetski D, Blum D, Guiel T, Heaton R, Levine J, Akoulitchev A. Development and Validation of Blood-Based Predictive Biomarkers for Response to PD-1/PD-L1 Checkpoint Inhibitors: Evidence of a Universal Systemic Core of 3D Immunogenetic Profiling across Multiple Oncological Indications. Cancers (Basel) 2023; 15:2696. [PMID: 37345033 DOI: 10.3390/cancers15102696] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/06/2023] [Accepted: 05/08/2023] [Indexed: 06/23/2023] Open
Abstract
BACKGROUND Unprecedented advantages in cancer treatment with immune checkpoint inhibitors (ICIs) remain limited to only a subset of patients. Systemic analyses of the regulatory 3D genome architecture linked to individual epigenetic and immunogenetic controls associated with tumour immune evasion mechanisms and immune checkpoint pathways reveal a highly prevalent molecular profile predictive of response to PD-1/PD-L1 ICIs. A clinical blood test based on a set of eight (8) 3D genomic biomarkers has been developed and validated on the basis of an observational trial to predict response to ICI therapy. METHODS The predictive eight biomarker set is derived from prospective observational clinical trials, representing 280 treatments with Pembrolizumab, Atezolizumab, Durvalumab, Nivolumab, and Avelumab in a broad range of indications: melanoma, lung, hepatocellular, renal, breast, bladder, colon, head and neck, bone, brain, lymphoma, prostate, vulvar, and cervical cancers. RESULTS The 3D genomic eight biomarker panel for response to immune checkpoint therapy achieved a high accuracy of 85%, sensitivity of 93%, and specificity of 82%. CONCLUSIONS This study demonstrates that a 3D genomic approach can be used to develop a predictive clinical assay for response to PD-1/PD-L1 checkpoint inhibition in cancer patients.
Collapse
Affiliation(s)
| | | | | | - Ann Dring
- Oxford BioDynamics Plc., Oxford OX4 2WB, UK
| | | | | | | | - Mutaz Issa
- Oxford BioDynamics Plc., Oxford OX4 2WB, UK
| | | | - Serene Ng
- Oxford BioDynamics (M) Sdn Bhd, Penang 10470, Malaysia
| | - Chun Ren Lim
- Oxford BioDynamics (M) Sdn Bhd, Penang 10470, Malaysia
| | - Cheah Soon Keat
- Mount Miriam Cancer Hospital (MMCH), Penang 11200, Malaysia
- Island Hospital, Penang 10450, Malaysia
| | - Ang Tick Suan
- Mount Miriam Cancer Hospital (MMCH), Penang 11200, Malaysia
| | - Rakesh Raman
- Mount Miriam Cancer Hospital (MMCH), Penang 11200, Malaysia
| | - Ho Kean Fatt
- Mount Miriam Cancer Hospital (MMCH), Penang 11200, Malaysia
| | | | - Heba Alshaker
- School of Medicine, University of East Anglia, Norwich NR4 7TJ, UK
| | | | - Dave Blum
- Oxford BioDynamics Inc., Gaithersburg, MD 20878, USA
| | - Thomas Guiel
- Oxford BioDynamics Inc., Gaithersburg, MD 20878, USA
| | - Robert Heaton
- Oxford BioDynamics Inc., Gaithersburg, MD 20878, USA
| | - Jedd Levine
- Oxford BioDynamics Inc., Gaithersburg, MD 20878, USA
| | | |
Collapse
|
8
|
Hattori M, Masuda N, Takano T, Tsugawa K, Inoue K, Matsumoto K, Ishikawa T, Itoh M, Yasojima H, Tanabe Y, Yamamoto K, Suzuki M, Pan W, Cortes J, Iwata H. Pembrolizumab plus chemotherapy in Japanese patients with triple-negative breast cancer: Results from KEYNOTE-355. Cancer Med 2023; 12:10280-10293. [PMID: 36916728 DOI: 10.1002/cam4.5757] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/10/2023] [Accepted: 02/20/2023] [Indexed: 03/16/2023] Open
Abstract
Pembrolizumab plus chemotherapy improved progression-free survival (PFS) and overall survival (OS) compared with placebo plus chemotherapy in patients with previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer with tumor programmed cell death ligand 1 (PD-L1) combined positive score (CPS) ≥10 in the global, phase 3, randomized controlled trial KEYNOTE-355. We report results for patients enrolled in Japan. Patients were randomized 2:1 to pembrolizumab 200 mg or placebo Q3W for 35 cycles plus chemotherapy (nab-paclitaxel, paclitaxel, or gemcitabine-carboplatin). Primary endpoints were PFS per RECIST version 1.1 by blinded independent central review and OS in patients with PD-L1 CPS ≥10, PD-L1 CPS ≥1, and the intention-to-treat (ITT) population. No alpha was assigned to this exploratory analysis. Eighty-seven patients were randomized in Japan (pembrolizumab plus chemotherapy, n = 61; placebo plus chemotherapy, n = 26), 66 (76%) had PD-L1 CPS ≥1, and 28 (32%) had PD-L1 CPS ≥10. Median time from randomization to data cutoff (June 15, 2021) was 44.7 (range, 37.2-52.9) months in the ITT population. Hazard ratios (HRs; 95% CI) for OS were 0.36 (0.14-0.89), 0.52 (0.30-0.91), and 0.46 (0.28-0.77) in the PD-L1 CPS ≥10, PD-L1 CPS ≥1, and ITT populations, respectively. HRs (95% CI) for PFS were 0.52 (0.20-1.34), 0.61 (0.35-1.06), and 0.64 (0.39-1.05). Grade 3 or 4 treatment-related adverse events occurred in 85% of patients in each group (no grade 5 events). Consistent with the global population, pembrolizumab plus chemotherapy tended to show improvements in OS and PFS with manageable toxicity versus placebo plus chemotherapy in Japanese patients and supports this combination in this setting.
Collapse
Affiliation(s)
| | - Norikazu Masuda
- Nagoya University Graduate School of Medicine, Nagoya, Japan.,National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Toshimi Takano
- The Cancer Institute Hospital of JFCR, Tokyo, Japan.,Toranomon Hospital, Tokyo, Japan
| | | | | | | | | | - Mitsuya Itoh
- Hiroshima City Hiroshima Citizens Hospital, Hiroshima, Japan
| | - Hiroyuki Yasojima
- National Hospital Organization Osaka National Hospital, Osaka, Japan
| | | | | | | | - Wilbur Pan
- Merck & Co., Inc., Rahway, New Jersey, USA
| | - Javier Cortes
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quironsalud Group, Madrid and Barcelona, Spain and Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid, Spain
| | | |
Collapse
|
9
|
Goudy OJ, Peng A, Tripathy A, Kuhlman B. Design of a protease-activated PD-L1 inhibitor. Protein Sci 2023; 32:e4578. [PMID: 36705186 PMCID: PMC9926466 DOI: 10.1002/pro.4578] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/18/2023] [Accepted: 01/25/2023] [Indexed: 01/28/2023]
Abstract
Immune checkpoint inhibitors that bind to the cell surface receptor PD-L1 are effective anti-cancer agents but suffer from immune-related adverse events as PD-L1 is expressed on both healthy and cancer cells. To mitigate toxicity, researchers are testing prodrugs that have low affinity for checkpoint targets until activated with proteases enriched in the tumor microenvironment. Here, we engineer a prodrug form of a PD-L1 inhibitor. The inhibitor is a soluble PD-1 mimetic that was previously engineered to have high affinity for PD-L1. In the basal state, the binding surface of the PD-1 mimetic is masked by fusing it to a soluble variant of its natural ligand, PD-L1. Proteolytic cleavage of the linker that connects the mask to the inhibitor activates the molecule. To optimize the mask so that it effectively blocks binding to PD-L1 but releases upon cleavage, we tested a set of mutants with varied affinity for the inhibitor. The top-performing mask reduces the affinity of the prodrug for PD-L1 120-fold, and binding is nearly fully recovered upon cleavage. In a cell-based assay measuring inhibition of the PD-1:PD-L1 interaction on the surface of cells, the IC50s of the masked inhibitors were up to 40-fold higher than their protease-treated counterparts. The changes in activity we observe upon protease treatment are comparable to systems currently tested in the clinic and provide evidence that natural binding partners are an excellent starting point for creating a prodrug.
Collapse
Affiliation(s)
- Odessa J. Goudy
- Department of Biochemistry and BiophysicsUniversity of North Carolina School of MedicineChapel HillNorth CarolinaUSA
| | - Alice Peng
- Department of Biochemistry and BiophysicsUniversity of North Carolina School of MedicineChapel HillNorth CarolinaUSA
| | - Ashutosh Tripathy
- Department of Biochemistry and BiophysicsUniversity of North Carolina School of MedicineChapel HillNorth CarolinaUSA
| | - Brian Kuhlman
- Department of Biochemistry and BiophysicsUniversity of North Carolina School of MedicineChapel HillNorth CarolinaUSA
- Lineberger Comprehensive Cancer CenterUniversity of North CarolinaChapel HillNorth CarolinaUSA
| |
Collapse
|
10
|
Cai H, Ren Y, Chen S, Wang Y, Chu L. Ferroptosis and tumor immunotherapy: A promising combination therapy for tumors. Front Oncol 2023; 13:1119369. [PMID: 36845720 PMCID: PMC9945274 DOI: 10.3389/fonc.2023.1119369] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 01/24/2023] [Indexed: 02/11/2023] Open
Abstract
Low response rate and treatment resistance are frequent problems in the immunotherapy of tumors, resulting in the unsatisfactory therapeutic effects. Ferroptosis is a form of cell death characterized by the accumulation of lipid peroxides. In recent years, it has been found that ferroptosis may be related to the treatment of cancer. Various immune cells (including macrophages and CD8+ T cells) can induce ferroptosis of tumor cells, and synergistically enhance the anti-tumor immune effects. However, the mechanisms are different for each cell types. DAMP released in vitro by cancer cells undergoing ferroptosis lead to the maturation of dendritic cells, cross-induction of CD8+ T cells, IFN-γ production and M1 macrophage production. Thus, it activates the adaptability of the tumor microenvironment and forms positive feedback of the immune response. It suggests that induction of ferroptosis may contribute to reducing resistance of cancer immunotherapy and has great potential in cancer therapy. Further research into the link between ferroptosis and tumor immunotherapy may offer hope for those cancers that are difficult to treat. In this review, we focus on the role of ferroptosis in tumor immunotherapy, explore the role of ferroptosis in various immune cells, and discuss potential applications of ferroptosis in tumor immunotherapy.
Collapse
Affiliation(s)
- Huazhong Cai
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, China,*Correspondence: Huazhong Cai,
| | - Yongfei Ren
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Shuangwei Chen
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yue Wang
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Liangmei Chu
- Department of Radiation Oncology, Institute of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
11
|
Wang Q, Li G, Ma X, Liu L, Liu J, Yin Y, Li H, Chen Y, Zhang X, Zhang L, Sun L, Ai J, Xu S. LncRNA TINCR impairs the efficacy of immunotherapy against breast cancer by recruiting DNMT1 and downregulating MiR-199a-5p via the STAT1-TINCR-USP20-PD-L1 axis. Cell Death Dis 2023; 14:76. [PMID: 36725842 PMCID: PMC9892521 DOI: 10.1038/s41419-023-05609-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 02/03/2023]
Abstract
Although programmed death-ligand 1 (PD-L1) inhibitors have achieved some therapeutic success in breast cancer, their efficacy is limited by low therapeutic response rates, which is closely related to the immune escape of breast cancer cells. Tissue differentiation inducing non-protein coding RNA (TINCR), a long non-coding RNA, as an oncogenic gene associated with the progression of various malignant tumors, including breast cancer; however, the role of TINCR in tumor immunity, especially in breast cancer, remains unclear. We confirmed that TINCR upregulated PD-L1 expression in vivo and in vitro, and promoted the progression of breast cancer. Next, we revealed that TINCR knockdown can significantly improve the therapeutic effect of PD-L1 inhibitors in breast cancer in vivo. Mechanistically, TINCR recruits DNMT1 to promote the methylation of miR-199a-5p loci and inhibit its transcription. Furthermore, in the cytoplasm, TINCR potentially acts as a molecular sponge of miR-199a-5p and upregulates the stability of USP20 mRNA through a competing endogenous RNA (ceRNA) regulatory mechanism, thus promoting PD-L1 expression by decreasing its ubiquitination level. IFN-γ stimulation activates STAT1 by phosphorylation, which migrates into the nucleus to promote TINCR transcription. This is the first study to describe the regulatory role of TINCR in breast cancer tumor immunity, broadening the current paradigm of the functional diversity of TINCR in tumor biology. In addition, our study provides new research directions and potential therapeutic targets for PD-L1 inhibitors in breast cancer.
Collapse
Affiliation(s)
- Qin Wang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy of Harbin Medical University, 157 Baojian Road, 150086, Harbin, China
- Sino-Russian Medical Research Center, Harbin Medical University Cancer Hospital, 150 Haping Road, 150081, Harbin, China
- Heilongjiang Academy of Medical Sciences, 157 Baojian Road, 150086, Harbin, China
| | - Guozheng Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, 150040, Harbin, China
| | - Xin Ma
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, 150040, Harbin, China
| | - Lei Liu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, 150040, Harbin, China
| | - Jiena Liu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, 150040, Harbin, China
| | - Yanling Yin
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, 150040, Harbin, China
| | - Hui Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, 150040, Harbin, China
| | - Yihai Chen
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, 150040, Harbin, China
| | - Xin Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, 150040, Harbin, China
| | - Lei Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, 150040, Harbin, China
| | - Liyang Sun
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy of Harbin Medical University, 157 Baojian Road, 150086, Harbin, China
| | - Jing Ai
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy of Harbin Medical University, 157 Baojian Road, 150086, Harbin, China.
| | - Shouping Xu
- Heilongjiang Academy of Medical Sciences, 157 Baojian Road, 150086, Harbin, China.
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, 150040, Harbin, China.
| |
Collapse
|
12
|
PD-L1: expression regulation. BLOOD SCIENCE 2023; 5:77-91. [DOI: 10.1097/bs9.0000000000000149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/29/2022] [Indexed: 02/05/2023] Open
|
13
|
Alimohammadi M, Faramarzi F, Mafi A, Mousavi T, Rahimi A, Mirzaei H, Asemi Z. Efficacy and Safety of Atezolizumab Monotherapy or Combined Therapy with Chemotherapy in Patients with Metastatic Triple-negative Breast Cancer: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Curr Pharm Des 2023; 29:2461-2476. [PMID: 37921135 DOI: 10.2174/0113816128270102231016110637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/16/2023] [Accepted: 09/21/2023] [Indexed: 11/04/2023]
Abstract
INTRODUCTION Several successful attempts have been recorded with PD-L1 blockade via atezolizumab monotherapy or combination therapy with chemotherapy in patients with metastatic triple-negative breast cancer (mTNBC). Due to the lack of a large-scale study, we present a meta-analysis aimed at evaluating the safety and efficacy of this promising strategy in patients with mTNBC. METHODS A comprehensive literature search was conducted using electronic databases to identify eligible RCTs. Twelve studies, including 2479 mTBNC patients treated with atezolizumab monotherapy or in combination with chemotherapy, were included up to January 2022. The PRISMA checklist protocol and the I2 statistic were applied for quality assessment and heterogeneity tests of the selected trials, respectively. Fixed and random-effects models were estimated based on the heterogeneity tests, and statistical analysis was performed using CMA. RESULTS Our pooled findings demonstrated that the median overall survival (OS) and progression-free survival (PFS) were 16.526 and 5.814 months in mTNBC patients, respectively. Furthermore, when comparing efficacy indicators between PD-L1-positive and PD-L1-negative groups, mTNBC patients with PD-L1 had better OS, PFS, and ORR than PD-L1-negative patients. Also, the immune-related adverse event incident for alopecia was higher (51.9%) than other complications across atezolizumab therapy. CONCLUSION Moreover, the pooled analysis indicated that the overall rate of lung metastasis following atezolizumab therapy was 42.8%, which was higher than the rates of metastasis in bone (26.9%), brain (5.4%), and lymph node (6.5%). Atezolizumab showed a manageable safety profile and had promising and durable anti-tumor efficacy in TMBC patients. Higher PD-L1 expression may be closely correlated with better clinical efficacy.
Collapse
Affiliation(s)
- Mina Alimohammadi
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Faramarzi
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Alireza Mafi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Tahoora Mousavi
- Molecular and Cell Biology Research Center (MCBRC), Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
- Medical Sciences Technologies, Molecular and Cell Biology Research Center (MCBRC), Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ali Rahimi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
14
|
Kenmotsu H, Sugawara S, Watanabe Y, Saito H, Okada M, Chen‐Yoshikawa TF, Ohe Y, Nishio W, Nakagawa S, Nagao H. Adjuvant atezolizumab in Japanese patients with resected stage IB-IIIA non-small cell lung cancer (IMpower010). Cancer Sci 2022; 113:4327-4338. [PMID: 36062851 PMCID: PMC9746048 DOI: 10.1111/cas.15564] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 08/25/2022] [Accepted: 08/28/2022] [Indexed: 12/15/2022] Open
Abstract
The global phase 3 IMpower010 study evaluated adjuvant atezolizumab versus best supportive care (BSC) following platinum-based chemotherapy in patients with resected stage IB-IIIA non-small cell lung cancer (NSCLC). Here, we report a subgroup analysis in patients enrolled in Japan. Eligible patients had complete resection of histologically or cytologically confirmed stage IB (tumors ≥4 cm)-IIIA NSCLC. Upon completing 1-4 cycles of adjuvant cisplatin-based chemotherapy, patients were randomized 1:1 to receive atezolizumab (fixed dose of 1200 mg every 21 days; 16 cycles or 1 year) or BSC. The primary endpoint of the global IMpower010 study was investigator-assessed disease-free survival, tested hierarchically first in patients with stage II-IIIA NSCLC whose tumors expressed programmed death-ligand 1 (PD-L1) on ≥1% of tumor cells, then in all randomized patients with stage II-IIIA NSCLC, and finally in the intention-to-treat (ITT) population (stage IB-IIIA NSCLC). Safety was evaluated in all patients who received atezolizumab or BSC. The study comprised 149 enrolled patients in three populations: ITT (n = 117; atezolizumab, n = 59; BSC, n = 58), all-randomized stage II-IIIA (n = 113; atezolizumab, n = 56; BSC, n = 57), and PD-L1 tumor cells ≥1% stage II-IIIA (n = 74; atezolizumab, n = 41; BSC, n = 33). At the data cutoff date (January 21, 2021), a trend toward disease-free survival improvement with atezolizumab vs BSC was observed in the PD-L1 tumor cells ≥1% stage II-IIIA (unstratified hazard ratio [HR], 0.52; 95% confidence interval [CI], 0.25-1.08), all-randomized stage II-IIIA (unstratified HR, 0.62; 95% CI, 0.35-1.11), and ITT (unstratified HR, 0.61; 95% CI, 0.34-1.10) populations. Atezolizumab-related grade 3/4 adverse events occurred in 16% of patients; no treatment-related grade 5 events occurred. Adjuvant atezolizumab showed disease-free survival improvement and a tolerable toxicity profile in Japanese patients in IMpower010, consistent with the global study results.
Collapse
Affiliation(s)
| | | | | | - Haruhiro Saito
- Department of Thoracic OncologyKanagawa Cancer CenterYokohamaJapan
| | - Morihito Okada
- Department of Surgical OncologyHiroshima UniversityHiroshimaJapan
| | | | | | - Wataru Nishio
- Department of Chest SurgeryHyogo Cancer CenterAkashiJapan
| | | | | |
Collapse
|
15
|
Recent advances in atezolizumab-based programmed death-ligand 1 (PD-L1) blockade therapy for breast cancer. Int Immunopharmacol 2022; 113:109334. [DOI: 10.1016/j.intimp.2022.109334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 10/08/2022] [Accepted: 10/08/2022] [Indexed: 11/05/2022]
|
16
|
Sharmni Vishnu K, Win TT, Aye SN, Basavaraj AK. Combined atezolizumab and nab-paclitaxel in the treatment of triple negative breast cancer: a meta-analysis on their efficacy and safety. BMC Cancer 2022; 22:1139. [PMID: 36335316 PMCID: PMC9637314 DOI: 10.1186/s12885-022-10225-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022] Open
Abstract
Background Triple negative breast cancer (TNBC) is clinically aggressive breast cancer with a poor prognosis. Approximately 20% of TNBC has been found to express programmed death ligand 1 (PD-L1), making it a potential therapeutic target. As a PD-L1 inhibitor, atezolizumab is a recently approved immunotherapeutic drug for TNBC, this meta-analysis (MA) was aimed to review the randomized controlled trial studies (RCTs) of combined atezolizumab and nab-paclitaxel in the treatment of TNBC and synthesize the evidence-based results on its effectiveness and safety. Method We searched PubMed, Embase, EBSCOhost and ClinicalTrials.gov for the eligible RCTs which compared the efficacy and safety of combined atezolizumab and nab-paclitaxel with nab-paclitaxel alone. The outcomes analyzed included overall survival (OS), progression-free survival (PFS), objective response rate (ORR) and treatment-related adverse effects (AEs). Results A total of six RCTs were included in this MA. For efficacy, although OS was not significantly prolonged with combined atezolizumab and nab-paclitaxel (HR 0.90, 95% CI [0.79, 1.01], p=0.08), this combination therapy significantly improved PFS (HR 0.72, 95% CI [0.59, 0.87], p=0.0006) and ORR (RR 1.25, 95% CI [0.79, 1.01] p<0.00001). For safety, any AEs, haematological, gastrointestinal, and liver AEs showed no statistically significant differences between the atezolizumab and nab-paclitaxel combination group and nab-paclitaxel alone group. However, serious AEs, high grade, dermatological, pulmonary, endocrine, and neurological AEs were significantly lower with nab-paclitaxel alone compared to atezolizumab and nab-paclitaxel combined (p-value range from <0.00001 to 0,02). Conclusion Atezolizumab combined with nab-paclitaxel was associated with improved outcomes in the treatment of TNBC; however, this combination resulted in more toxicity compared to nab-paclitaxel alone. While nab-paclitaxel alone produced chemotherapy-related AEs, the combination of atezolizumab with nab-paclitaxel produced AEs, especially immune-related AEs such as haematological, pulmonary, endocrine, and neurological AEs. Trial registration This research work of systematic review has been registered on PROSPERO (Registration number: CRD42022297952). Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-10225-y.
Collapse
|
17
|
Yuan K, Wu J, Zhao Y, Lyu S, Zhou Q, Shi F, Li Y, Song Q. Consistent expression of PD-L1 in tumor microenvironment with peripheral PD-1/PD-L1 in circulating T lymphocytes of operable breast cancer: a diagnostic test. Diagn Pathol 2022; 17:68. [PMID: 36088412 PMCID: PMC9464389 DOI: 10.1186/s13000-022-01249-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/19/2022] [Indexed: 12/24/2022] Open
Abstract
Background The expression of PD-L1 in the immune microenvironment can guide the application of immunosuppressants. In order to monitor the immune status of the body, repeated biopsies have to be taken. Our research aims to find new and convenient means to evaluate this indicator. Methods Eighty-three cases of newly diagnosed operable breast cancer without receiving preoperative treatment, were recruited from Beijing Shijitan Hospital between November 2018 and November 2019. The expression of PD-1/PD-L1 on circulating T lymphocytes was detected by flow cytometry and the expression of PD-L1 on immune cells in tumor microenvironment was detected by immunohistochemistry. Results The median percentage of positive PD-1 and PD-L1 expression on circulating T lymphocytes was 15.2% and 0.7%, respectively. The peripheral PD-1 had no relationship with clinicopathological characteristics, but the peripheral PD-L1 expression had a correlation with lymph node metastasis (p = 0.005) and Her-2 expression (p = 0.034) (p < 0.05). The positive rate of PD-L1 expression was 32.9% in tumor microenvironment. PD-L1 expression in tumor microenvironment had a significant correlation with PD-1/PD-L1 expression on circulating T lymphocytes, the correlation coefficients being 0.24 (p < 0.05) and 0.26 (p < 0.05), respectively. To predict the PD-L1 expression in tumor microenvironment, the area under the receiver operating characteristic curve was 0.65 and 0.66 for peripheral PD-1 and PD-L1, respectively. High level of peripheral PD-1/PD-L1 expression was associated with the odds ratios of 5.42 and 4.76 for positive PD-L1 expression in tumor microenvironment. Conclusion Peripheral PD-1/PD-L1 expression had a significant consistency with PD-L1 expression in tumor microenvironment and could act as an alternative choice of tissue detection, for the patients intolerable of biopsy.
Collapse
|
18
|
Wesolowski J, Tankiewicz-Kwedlo A, Pawlak D. Modern Immunotherapy in the Treatment of Triple-Negative Breast Cancer. Cancers (Basel) 2022; 14:cancers14163860. [PMID: 36010854 PMCID: PMC9406094 DOI: 10.3390/cancers14163860] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/04/2022] [Accepted: 08/07/2022] [Indexed: 11/29/2022] Open
Abstract
Simple Summary This review summarizes reports from the latest clinical trials assessing the safety and clinical effectiveness of new biological drugs stimulating the immune system to fight cancer. The aim of this study is to show the enormous therapeutic potential of monoclonal antibodies in the treatment of cancer, in particular triple negative breast cancer (TNBC). Introduction of these innovative drugs to the standard clinical cancer therapies, including TNBC, allows for an increase in the response rate to the applied treatment, and consequently extending the lives of patients suffering from cancer. We hope to draw attention to the extremely difficult-to-treat TNBC, as well as the importance of the development of clinical trials evaluating drugs modulating the immune system in TNBC therapy. Abstract Triple-Negative Breast Cancer is a subtype of breast cancer characterized by the lack of expression of estrogen receptors, progesterone receptors, as well as human epidermal growth factor receptor 2. This cancer accounts for 15–20% of all breast cancers and is especially common in patients under 40 years of age, as well as with the occurring BRCA1 mutation. Its poor prognosis is reflected in the statistical life expectancy of 8–15 months after diagnosis of metastatic TNBC. So far, the lack of targeted therapy has narrowed therapeutic possibilities to classic chemotherapy. The idea behind the use of humanized monoclonal antibodies, as inhibitors of immunosuppressive checkpoints used by the tumor to escape from immune system control, is to reduce immunotolerance and direct an intensified anti-tumor immune response. An abundance of recent studies has provided numerous pieces of evidence about the safety and clinical benefits of immunotherapy using humanized monoclonal antibodies in the fight against many types of cancer, including TNBC. In particular, phase three clinical trials, such as the IMpassion 130, the KEYNOTE-355 and the KEYNOTE-522 resulted in the approval of immunotherapeutic agents, such as atezolizumab and pembrolizumab by the US Food and Drug Administration in TNBC therapy. This review aims to present the huge potential of immunotherapy using monoclonal antibodies directed against immunosuppressive checkpoints—such as atezolizumab, avelumab, durvalumab, pembrolizumab, nivolumab, cemiplimab, tremelimumab, ipilimumab—in the fight against difficult to treat TNBCs as monotherapy as well as in more advanced combination strategies.
Collapse
Affiliation(s)
- Jakub Wesolowski
- Department of Pharmacodynamics, Faculty of Pharmacy, Medical University in Bialystok, 15-089 Bialystok, Poland
- Correspondence:
| | - Anna Tankiewicz-Kwedlo
- Department of Monitored Pharmacotherapy, Faculty of Pharmacy, Medical University in Bialystok, 15-089 Bialystok, Poland
| | - Dariusz Pawlak
- Department of Pharmacodynamics, Faculty of Pharmacy, Medical University in Bialystok, 15-089 Bialystok, Poland
| |
Collapse
|
19
|
Saji S, Ohsumi S, Ito M, Hayashi N, Kobayashi K, Masuda N, Niikura N, Yamashita T, Kiyama K, Hasegawa A, Nakagawa S, Hattori M. Subgroup analysis of Japanese patients in a phase III randomized, controlled study of neoadjuvant atezolizumab or placebo, combined with nab-paclitaxel and anthracycline-based chemotherapy in early triple-negative breast cancer (IMpassion031). Jpn J Clin Oncol 2022; 52:1124-1133. [PMID: 35750038 PMCID: PMC9538755 DOI: 10.1093/jjco/hyac098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/27/2022] [Indexed: 11/29/2022] Open
Abstract
Background In the global phase III IMpassion031 study, neoadjuvant atezolizumab plus nab-paclitaxel/anthracycline-based chemotherapy improved pathological complete response in patients with early stage triple-negative breast cancer. Here, we report primary analysis results from a subgroup of Japanese patients. Methods Patients with histologically documented, previously untreated, stage cT2–cT4, cN0–cN3, cM0 triple-negative breast cancer were randomized 1:1 to receive intravenous atezolizumab 840 mg or placebo every 2 weeks in combination with chemotherapy consisting of nab-paclitaxel intravenous 125 mg/m2 once a week, followed by doxorubicin intravenous 60 mg/m2 and cyclophosphamide intravenous 600 mg/m2 every 2 weeks. Patients then underwent surgery. Pathological complete response (ypT0/is ypN0) in the intention-to-treat and PD-L1-positive (≥1% PD-L1-expressing tumor-infiltrating immune cells) populations were co-primary endpoints. Results This subanalysis (data cutoff: 3 April 2020) included 36 patients from Japan (intention-to-treat; atezolizumab arm, n = 17; placebo arm, n = 19). Pathological complete response occurred in 41% (n = 7; 95% confidence interval, 18–67) of patients in the atezolizumab arm and 37% (n = 7; 95% confidence interval, 16–62) in the placebo arm. In the PD-L1-positive population, pathological complete response occurred in 50% (n = 5; 95% confidence interval, 19–81) of patients in the atezolizumab arm and 45% (n = 5; 95% confidence interval, 17–77) in the placebo arm. Treatment-related grade 3–4 adverse events occurred in 71% and 68% of patients in the respective arms. Conclusion Atezolizumab added to neoadjuvant chemotherapy numerically improved pathological complete response versus placebo in this small exploratory analysis of Japanese patients with early stage triple-negative breast cancer, a trend directionally consistent with the global study results. No new safety signals were identified.
Collapse
Affiliation(s)
- Shigehira Saji
- Department of Medical Oncology, Fukushima Medical University, Fukushima, Japan
| | - Shozo Ohsumi
- Department of Breast Oncology, National Hospital Organization Shikoku Cancer Center, Ehime, Japan
| | - Mitsuya Ito
- Department of Breast Surgery, Hiroshima City Hiroshima Citizens Hospital, Hiroshima, Japan
| | - Naoki Hayashi
- Department of Breast Surgical Oncology, St. Luke's International Hospital, Tokyo, Japan
| | - Kokoro Kobayashi
- Department of Medical Oncology, The Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Norikazu Masuda
- Department of Surgery, Breast Oncology, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Naoki Niikura
- Department of Breast and Endocrine Surgery, Tokai University School of Medicine, Kanagawa, Japan
| | - Toshinari Yamashita
- Department of Breast and Endocrine Surgery, Kanagawa Cancer Center, Kanagawa, Japan
| | | | | | | | - Masaya Hattori
- Department of Breast Oncology, Aichi Cancer Center Hospital, Aichi, Japan
| |
Collapse
|
20
|
Steiger HJ, Vollmer K, Rogers S, Schwyzer L. State of affairs regarding targeted pharmacological therapy of cancers metastasized to the brain. Neurosurg Rev 2022; 45:3119-3138. [PMID: 35902427 PMCID: PMC9492578 DOI: 10.1007/s10143-022-01839-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 02/03/2023]
Abstract
In 1999 a visionary short article by The Wall Street Journal writers Robert Langreth and Michael Waldholz popularized the new term "personalized medicine," that is to say, the targeting of drugs to each unique genetic profile. From today's perspective, targeted approaches have clearly found the widest use in the antineoplastic domain. The current review was initiated to review the progress that has been made regarding the treatment of patients with advanced cancer and brain metastases. PubMed was searched for the terms brain metastasis, brain metastases, or metastatic brain in the Title/Abstract. Selection was limited to randomized controlled trial (RCT) and publication date January 2010 to February 2022. Following visual review, 51 papers on metastatic lung cancer, 12 on metastatic breast cancer, and 9 on malignant melanoma were retained and underwent full analysis. Information was extracted from the papers giving specific numbers for intracranial response rate and/or overall survival. Since most pharmacological trials on advanced cancers excluded patients with brain metastases and since hardly any information on adjuvant radiotherapy and radiosurgery is available from the pharmacological trials, precise assessment of the effect of targeted medication for the subgroups with brain metastases is difficult. Some quantitative information regarding the success of targeted pharmacological therapy is only available for patients with breast and lung cancer and melanoma. Overall, targeted approaches approximately doubled the lifespan in the subgroups of brain metastases from tumors with targetable surface receptors such as anaplastic lymphoma kinase (ALK) fusion receptor in non-small cell lung cancer or human epidermal growth factor receptor 2 (HER2)-positive breast cancer. For these types, overall survival in the situation of brain metastases is now more than a year. For receptor-negative lung cancer and melanoma, introduction of immune checkpoint blockers brought a substantial advance, although overall survival for melanoma metastasized to the brain appears to remain in the range of 6 to 9 months. The outlook for small cell lung cancer metastasized to the brain apparently remains poor. The introduction of targeted therapy roughly doubled survival times of advanced cancers including those metastasized to the brain, but so far, targeted therapy does not differ essentially from chemotherapy, therefore also facing tumors developing escape mechanisms. With the improved perspective of patients suffering from brain metastases, it becomes important to further optimize treatment of this specific patient group within the framework of randomized trials.
Collapse
Affiliation(s)
- Hans-Jakob Steiger
- Department of Neurosurgery, Kantonsspital Aarau, Aarau, Switzerland.
- Klinik Für Neurochirurgie, Neurozentrum, Kantonsspital Aarau, Tellstr. 25, CH-5001, Aarau, Switzerland.
- Department of Neurosurgery, University Hospital Düsseldorf, Düsseldorf, Germany.
| | - Kathrin Vollmer
- Division of Oncology, Hematology and Transfusion Medicine, Kantonsspital Aarau, Aarau, Switzerland
| | - Susanne Rogers
- Radio-Oncology-Centre KSA-KSB, Kantonsspital Aarau, Aarau, Switzerland
| | - Lucia Schwyzer
- Department of Neurosurgery, Kantonsspital Aarau, Aarau, Switzerland
| |
Collapse
|
21
|
Lu YS, Yeo W, Yap YS, Park YH, Tamura K, Li H, Cheng R. An Overview of the Treatment Efficacy and Side Effect Profile of Pharmacological Therapies in Asian Patients with Breast Cancer. Target Oncol 2021; 16:701-741. [PMID: 34582007 PMCID: PMC8613101 DOI: 10.1007/s11523-021-00838-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2021] [Indexed: 11/05/2022]
Abstract
Breast cancer (BC) among Asians accounts for ~ 40% of the global BC burden. Differences in BC risk, presentation, tumor biology, and response to treatment exist between Asian and non-Asian patients; however, Asian patients are often under-represented in clinical trials. This narrative review summarizes the efficacy and safety of pharmacological therapies for BC in Asian populations, with a focus on outcomes in Asian versus non-Asian patients treated with chemotherapy, hormone therapy, anti-human epidermal growth factor receptor-2 targeted therapies, cyclin-dependent kinase 4/6 (CDK4/6) inhibitors, mammalian target of rapamycin inhibitors, bone-targeted therapies, poly-ADP ribose polymerase, phosphoinositide 3-kinase, and checkpoint inhibitors. While most therapies have demonstrated comparable efficacy and safety in Asian and non-Asian patients with BC, differences that are largely attributed to pharmacogenetic variations between populations exist. Pharmacogenetic differences may contribute to a reduced clinical benefit of tamoxifen, whereas improved clinical outcomes have been reported with tyrosine kinase inhibitors and CDK4/6 inhibitors in Asian versus non-Asian patients with BC. In particular, Asian patients have an increased incidence of hematological toxicities, including neutropenia, although adverse events can be effectively managed using dose adjustments. Recent trials with CDK4/6 inhibitors have increased efforts to include Asians within study subsets. Future clinical trials enrolling higher numbers of Asian patients, and an increased understanding of differences in patient and tumor genetics between Asians and non-Asians, have the potential to incrementally improve the management of BC in Asian patients.
Collapse
Affiliation(s)
- Yen-Shen Lu
- Department of Oncology, National Taiwan University Hospital, No. 7, Chung Shan South Road, Taipei, 100, Taiwan.
| | - Winnie Yeo
- Department of Clinical Oncology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Yoon-Sim Yap
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Yeon Hee Park
- Sungkyunkwan University, SKKU-Samsung Medical Centre, Seoul, South Korea
| | - Kenji Tamura
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Huiping Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | | |
Collapse
|
22
|
Masoumi E, Tahaghoghi-Hajghorbani S, Jafarzadeh L, Sanaei MJ, Pourbagheri-Sigaroodi A, Bashash D. The application of immune checkpoint blockade in breast cancer and the emerging role of nanoparticle. J Control Release 2021; 340:168-187. [PMID: 34743998 DOI: 10.1016/j.jconrel.2021.10.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 12/13/2022]
Abstract
Breast cancer is the most common malignancy in the female population with a high mortality rate. Despite the satisfying depth of studies evaluating the contributory role of immune checkpoints in this malignancy, few articles have reviewed the pros and cons of immune checkpoint blockades (ICBs). In the current review, we provide an overview of immune-related inhibitory molecules and also discuss the original data obtained from international research laboratories on the aberrant expression of T and non-T cell-associated immune checkpoints in breast cancer. Then, we especially focus on recent studies that utilized ICBs as the treatment strategy in breast cancer and provide their efficiency reports. As there are always costs and benefits, we discuss the limitations and challenges toward ICB therapy such as adverse events and drug resistance. In the last section, we allocate an overview of the recent data concerning the application of nanoparticle systems for cancer immunotherapy and propose that nano-based ICB approaches may overcome the challenges related to ICB therapy in breast cancer. In conclusion, it seems it is time for nanoscience to more rapidly move forward into clinical trials and illuminates the breast cancer treatment area with its potent features for the target delivery of ICBs.
Collapse
Affiliation(s)
- Elham Masoumi
- Department of Immunology, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran; Student Research Committee, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Sahar Tahaghoghi-Hajghorbani
- Microbiology and Virology Research Center, Qaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Leila Jafarzadeh
- Department of Laboratory Science, Sirjan Faculty of Medical Science, Sirjan, Iran
| | - Mohammad-Javad Sanaei
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atieh Pourbagheri-Sigaroodi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
23
|
Morad G, Helmink BA, Sharma P, Wargo JA. Hallmarks of response, resistance, and toxicity to immune checkpoint blockade. Cell 2021; 184:5309-5337. [PMID: 34624224 DOI: 10.1016/j.cell.2021.09.020] [Citation(s) in RCA: 824] [Impact Index Per Article: 206.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/21/2021] [Accepted: 09/13/2021] [Indexed: 12/16/2022]
Abstract
Unprecedented advances have been made in cancer treatment with the use of immune checkpoint blockade (ICB). However, responses are limited to a subset of patients, and immune-related adverse events (irAEs) can be problematic, requiring treatment discontinuation. Iterative insights into factors intrinsic and extrinsic to the host that impact ICB response and toxicity are critically needed. Our understanding of the impact of host-intrinsic factors (such as the host genome, epigenome, and immunity) has evolved substantially over the past decade, with greater insights on these factors and on tumor and immune co-evolution. Additionally, we are beginning to understand the impact of acute and cumulative exposures-both internal and external to the host (i.e., the exposome)-on host physiology and response to treatment. Together these represent the current day hallmarks of response, resistance, and toxicity to ICB. Opportunities built on these hallmarks are duly warranted.
Collapse
Affiliation(s)
- Golnaz Morad
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Beth A Helmink
- Department of Surgery, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Padmanee Sharma
- Department of Genitourinary Medical Oncology and Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jennifer A Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
24
|
Balibegloo M, Nejadghaderi SA, Sadeghalvad M, Soleymanitabar A, Salehi Nezamabadi S, Saghazadeh A, Rezaei N. Adverse events associated with immune checkpoint inhibitors in patients with breast cancer: A systematic review and meta-analysis. Int Immunopharmacol 2021; 96:107796. [PMID: 34162158 DOI: 10.1016/j.intimp.2021.107796] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/15/2021] [Accepted: 05/15/2021] [Indexed: 01/10/2023]
Abstract
BACKGROUND Breast cancer is one of the most prevalent cancers with a high rate of mortality. Immune checkpoint inhibitors (ICIs) have shown promising results in breast cancer treatment. However, the incidences of adverse events (AEs) and immune-related AEs (irAEs) due to ICIs have not been investigated comprehensively. We aimed to determine any-grade and grade 3-5 AEs and irAEs of ICIs compared to the control group which were other conventional therapies in adults with breast cancer. METHODS We included controlled clinical trials that involved ICIs in adults with breast cancer to assess AEs and irAEs of ICIs. We systematically searched PubMed, EMBASE, Scopus, Web of Science, the Cochrane Central Register of Controlled Trials, Clinical-Trials.gov, and metaRegister of Controlled Trials up to March 12, 2021. Version 2 of the Cochrane risk-of-bias tool for randomized trials (RoB 2) was used for quality assessment. RESULTS Nine studies, including 4687 participants met our inclusion criteria. Rash and infusion reaction were the two most frequent irAEs of any-grade and grade 3-5. Among irAEs, hyperthyroidism had the most prominent difference between the two groups in favor of ICIs followed by hypothyroidism and adrenal insufficiency. Among grade 3-5 and any-grade non-immune AEs, increased aspartate aminotransferase (RR = 1.91; 95% CI, 1.11-3.28) and cough (RR = 1.32; 95%CI, 1.11-1.57) had the highest RRs in favor of ICIs, respectively. The frequencies of overall any-grade and grade 3-5 irAEs were higher in the ICI group. CONCLUSION The results showed that all the AEs and irAEs of all the categories were more prevalent with ICIs.
Collapse
Affiliation(s)
- Maryam Balibegloo
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Seyed Aria Nejadghaderi
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran; School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mona Sadeghalvad
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Soleymanitabar
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Student Research Committee, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Sasan Salehi Nezamabadi
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran; School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amene Saghazadeh
- Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
25
|
Ouyang T, Cao Y, Kan X, Chen L, Ren Y, Sun T, Yan L, Xiong B, Liang B, Zheng C. Treatment-Related Serious Adverse Events of Immune Checkpoint Inhibitors in Clinical Trials: A Systematic Review. Front Oncol 2021; 11:621639. [PMID: 34046338 PMCID: PMC8144509 DOI: 10.3389/fonc.2021.621639] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/19/2021] [Indexed: 12/11/2022] Open
Abstract
Background Immune Checkpoint Inhibitors (ICI) have been progressively used in cancer treatment and produced unique toxicity profiles. This systematic review aims to comprehend the patterns and occurrence of treatment-related adverse events (trAEs) based on ICI. Methods PICOS/PRISMA methods were used to identify published English-language on PubMed, Web of Science, and Scopus from 2015 to 2020. Published clinical trials on ICI monotherapy, combined ICIs, and ICI plus other treatment with tabulated data on grade≥3 trAEs were included. Odds ratio (OR), χ2 tests were used to analyze for effect size and associations. Results This review included 145 clinical trials involving 21786 patients. Grade 3-5 trAEs were more common with ICI when they were plused with other treatments compared with ICI monotherapy(54.3% versus 17.7%, 46.1%, p<0.05). Grade 3-5 trAEs were also more common with CTLA-4 mAbs compared with anti-PD-1 and anti-PD-L1 (34.2% versus 15.1%, 13.6%, p<0.05). Hyperthyroidism (OR 3.8, 95%CI 1.7–8.6), nausea (OR 3.7, 95%CI 2.5–5.3), diarrhea (OR 2.7, 95%CI 2.2–3.2), colitis (OR 3.4, 95%CI 2.7–4.3), ALT increase (OR 4.9, 95%CI 3.9–6.1), AST increase (OR 3.8, 95%CI 3.0–4.9), pruritus (OR 2.4, 95%CI 1.5–3.9), rash (OR 2.8, 95%CI 2.1–3.8), fatigue (OR 2.8, 95%CI 2.2–3.7), decreased appetite (OR 2.4, 95%CI 1.5–3.8), and hypophysitis (OR 2.0, 95%CI 1.2–3.3) were more frequent with combined ICIs. Diarrhea (OR 8.1, 95%CI 6.4–10.3), colitis (OR 12.2, 95%CI 8.7–17.1), ALT increase (OR 5.1, 95%CI 3.5–7.4), AST increase (OR 4.2, 95%CI 2.8–6.3), pruritus (OR 4.1, 95%CI 2.0–8.4), rash (OR 4.4, 95%CI 2.9–6.8), hypophysitis (OR 12.1, 95%CI 6.3–23.4) were more common with CTLA-4 mAbs; whereas pneumonitis (OR 4.7, 95% CI 2.1–10.3) were more frequent with PD-1 mAbs. Conclusions Different immune checkpoint inhibitors are associated with different treatment-related adverse events profiles. A comprehensive data in this systematic review will provide comprehensive information for clinicians.
Collapse
Affiliation(s)
- Tao Ouyang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Yanyan Cao
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Xuefeng Kan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Lei Chen
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Yanqiao Ren
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Tao Sun
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Liangliang Yan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Bin Xiong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Bin Liang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| |
Collapse
|
26
|
Jalalvand M, Darbeheshti F, Rezaei N. Immune checkpoint inhibitors: review of the existing evidence and challenges in breast cancer. Immunotherapy 2021; 13:587-603. [PMID: 33775102 DOI: 10.2217/imt-2020-0283] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cancer initiation and progression are associated with immune system responses. Tumor cells use various tricks to scape of immune system, such as activating immune checkpoint pathways that induce immunosuppressive functions. Among the different immune checkpoint receptors, CTLA-4 and PD-1/PD-L1 are prominent therapeutic targets in different cancers. Although the US FDA has approved some immune checkpoint inhibitors for several cancers, concerning breast cancer still different clinical trials are looking for optimizing efficacy and decreasing immune-related adverse events. This review will discuss the existing body of knowledge with regard to cross-talk between immune system and tumor cells and then explore immune checkpoint-related signaling pathways in the context of breast tumors. Finally, we highlight the application of different immune checkpoint blockers in breast cancer patients.
Collapse
Affiliation(s)
- Mobina Jalalvand
- Cancer Immunology Project (CIP), Universal Scientific Education & Research Network (USERN), Tehran, Iran.,School of medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzaneh Darbeheshti
- Department of Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Medical Genetics Network (MeGeNe), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran 14194, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 14194, Iran.,Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran 14194, Iran
| |
Collapse
|
27
|
Chisaki Y, Kuwada Y, Matsumura C, Yano Y. Cost-effectiveness Analysis of Atezolizumab Plus Nab-Paclitaxel for Advanced PD-L1 Positive Triple-Negative Breast Cancer in Japan. Clin Drug Investig 2021; 41:381-389. [PMID: 33674955 DOI: 10.1007/s40261-021-01017-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND AND OBJECTIVES Atezolizumab is an anti-programmed death ligand 1 (PD-L1) antibody that shows good safety and efficacy for patients with PD-L1-positive triple-negative breast cancer (TNBC). The cost of atezolizumab therapy is expensive, and its economic burden is an important problem. In this study, we evaluated the cost effectiveness of atezolizumab plus nab-paclitaxel therapy (AnP) compared with nab-paclitaxel monotherapy (nP) for PD-L1-positive TNBC under Japanese medical conditions and environments using a Markov model. METHODS The medical information was collected from data published by the IMpassion130 trial. A Markov model was established to simulate the number of patients in each disease state after AnP or nP therapy during each time period. As indices for cost effectiveness, total cost, quality-adjusted life years (QALYs), and incremental cost-effectiveness ratios (ICER) were calculated. Probabilistic sensitivity analysis (PSA) was used to assess the uncertainty of the model using 10,000 Monte Carlo simulations with difference parameters. RESULTS The QALYs for AnP treatment were longer than for nP treatment (1.12 vs 0.82 QALYs), but the total cost of AnP treatment was more expensive than that of nP treatment (¥11,070,143 vs ¥2,056,164). The ICER values comparing AnP treatment with nP treatment were ¥30,208,143/QALY. The ICER/QALY was more expensive than the willingness-to-pay (WTP) of ¥15,000,000 per QALY. To achieve a 50% cost-effective probability with a WTP threshold, the price of the atezolizumab should be reduced by 55.1%. CONCLUSIONS AnP was not cost effective compared to nP for PD-L1-positive inoperable TNBC under the Japanese condition.
Collapse
Affiliation(s)
- Yugo Chisaki
- Education and Research Center for Clinical Pharmacy, Kyoto Pharmaceutical University, 5-Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto, 607-8414, Japan.
| | - Yoshiki Kuwada
- Education and Research Center for Clinical Pharmacy, Kyoto Pharmaceutical University, 5-Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto, 607-8414, Japan
| | - Chikako Matsumura
- Education and Research Center for Clinical Pharmacy, Kyoto Pharmaceutical University, 5-Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto, 607-8414, Japan
| | - Yoshitaka Yano
- Education and Research Center for Clinical Pharmacy, Kyoto Pharmaceutical University, 5-Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto, 607-8414, Japan
| |
Collapse
|
28
|
Wang H, Ma H, Sové RJ, Emens LA, Popel AS. Quantitative systems pharmacology model predictions for efficacy of atezolizumab and nab-paclitaxel in triple-negative breast cancer. J Immunother Cancer 2021; 9:jitc-2020-002100. [PMID: 33579739 PMCID: PMC7883871 DOI: 10.1136/jitc-2020-002100] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2021] [Indexed: 12/18/2022] Open
Abstract
Background Immune checkpoint blockade therapy has clearly shown clinical activity in patients with triple-negative breast cancer, but less than half of the patients benefit from the treatments. While a number of ongoing clinical trials are investigating different combinations of checkpoint inhibitors and chemotherapeutic agents, predictive biomarkers that identify patients most likely to benefit remains one of the major challenges. Here we present a modular quantitative systems pharmacology (QSP) platform for immuno-oncology that incorporates detailed mechanisms of immune–cancer cell interactions to make efficacy predictions and identify predictive biomarkers for treatments using atezolizumab and nab-paclitaxel. Methods A QSP model was developed based on published data of triple-negative breast cancer. With the model, we generated a virtual patient cohort to conduct in silico virtual clinical trials and make retrospective analyses of the pivotal IMpassion130 trial that led to the accelerated approval of atezolizumab and nab-paclitaxel for patients with programmed death-ligand 1 (PD-L1) positive triple-negative breast cancer. Available data from clinical trials were used for model calibration and validation. Results With the calibrated virtual patient cohort based on clinical data from the placebo comparator arm of the IMpassion130 trial, we made efficacy predictions and identified potential predictive biomarkers for the experimental arm of the trial using the proposed QSP model. The model predictions are consistent with clinically reported efficacy endpoints and correlated immune biomarkers. We further performed a series of virtual clinical trials to compare different doses and schedules of the two drugs for simulated therapeutic optimization. Conclusions This study provides a QSP platform, which can be used to generate virtual patient cohorts and conduct virtual clinical trials. Our findings demonstrate its potential for making efficacy predictions for immunotherapies and chemotherapies, identifying predictive biomarkers, and guiding future clinical trial designs.
Collapse
Affiliation(s)
- Hanwen Wang
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Huilin Ma
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Richard J Sové
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Leisha A Emens
- Department of Medicine, University of Pittsburgh Medical Center, Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, Maryland, USA.,Department of Oncology, Johns Hopkins Medicine Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA
| |
Collapse
|
29
|
Atezolizumab (in Combination with Nab-Paclitaxel): A Review in Advanced Triple-Negative Breast Cancer. Drugs 2020; 80:601-607. [PMID: 32248356 DOI: 10.1007/s40265-020-01295-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Atezolizumab (Tecentriq®), an immune checkpoint inhibitor against programmed death ligand 1 (PD-L1), is the first immunotherapy agent to be approved (for use in combination with nab-paclitaxel) in the USA, the EU (as first-line) and Japan for the treatment of advanced triple-negative breast cancer (TNBC). Approval was based on the results of the phase III IMpassion130 trial in patients with unresectable locally advanced or metastatic TNBC, in which atezolizumab plus nab-paclitaxel significantly prolonged progression-free survival (PFS) when compared to placebo plus nab-paclitaxel in the intent-to-treat (ITT) population and the PD-L1+ subgroup. Statistically significant overall survival (OS) benefits were not seen in two interim analyses and final OS data are awaited. The tolerability and safety profile of atezolizumab plus nab-paclitaxel was consistent with those of each individual drug. The most common treatment-related adverse events included neutropenia, peripheral neuropathy and reduced neutrophil count. Adverse events of special interest occurred with higher frequency in patients who received atezolizumab plus nab-paclitaxel than placebo plus nab-paclitaxel, and were mostly immune-related (e.g. immune-related rash, hypothyroidism and hepatitis). Health-related quality of life was not significantly impacted by the addition of atezolizumab to nab-paclitaxel therapy. Thus, atezolizumab plus nab-paclitaxel is a useful immunochemotherapy option for patients with unresectable locally advanced or metastatic TNBC, including those whose tumours have PD-L1 expression ≥ 1%.
Collapse
|
30
|
Affiliation(s)
- Hideo Kunitoh
- Department of Medical Oncology, Japanese Red Cross Medical Center, Tokyo, Japan
| |
Collapse
|
31
|
Kwapisz D. Pembrolizumab and atezolizumab in triple-negative breast cancer. Cancer Immunol Immunother 2020; 70:607-617. [PMID: 33015734 DOI: 10.1007/s00262-020-02736-z] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 09/25/2020] [Indexed: 12/23/2022]
Abstract
Triple-negative breast cancer (TNBC) is defined by a lack of expression of both estrogen (ER) and progesterone (PgR) receptors as well as human epidermal growth factor receptor 2 (HER2) and is associated with poor prognosis. Moreover, the systemic treatment options are limited. However, the TNBC is more likely than other breast cancer subtypes to benefit from immune checkpoint blockade therapy due to its higher immunogenicity, higher enrichment by tumour-infiltrating lymphocytes (TILs), and higher levels of programmed cell death ligand 1 (PD-L1) expression. Thus far, atezolizumab was approved in combination with nab-paclitaxel for patients with unresectable locally advanced or metastatic TNBC whose tumours express PD-L1. Currently, it seems that PD-L1-positive subgroup will potentially benefit the most from the immune checkpoint inhibitor (ICI) treatment. Moreover, it seems that better results are seen when an ICI is given as first-line treatment than when an ICI is given in later lines of treatment for advanced TNBC/metastatic TNBC. Recently, pembrolizumab has demonstrated promising results in early-stage TNBC what can lead in near future to its approval in (neo)adjuvant setting. This review summarizes the development and highlights recent advances of the atezolizumab and pembrolizumab in early and advanced/metastatic TNBC.
Collapse
Affiliation(s)
- Dorota Kwapisz
- Department of Immunology, Transplantology and Internal Diseases, University Clinical Center of the Medical University in Warsaw, Warsaw, Poland.
| |
Collapse
|
32
|
Kern R, Correa SC, Scandolara TB, Carla da Silva J, Pires BR, Panis C. Current advances in the diagnosis and personalized treatment of breast cancer: lessons from tumor biology. Per Med 2020; 17:399-420. [PMID: 32804054 DOI: 10.2217/pme-2020-0070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Breast cancer treatment has advanced enormously in the last decade. Most of this is due to advances reached in the knowledge regarding tumor biology, mainly in the field of diagnosis and treatment. This review brings information about how the genomics-based information contributed to advances in breast cancer diagnosis and prognosis perspective, as well as presents how tumor biology discoveries fostered the main therapeutic approaches available to treat such patients, based on a personalized point of view.
Collapse
Affiliation(s)
- Rodrigo Kern
- Laboratory of Tumor Biology, State University of West Paraná, Francisco Beltrão - Paraná 85601-970, Brazil.,Post-Graduation Program in Health-Applied Sciences, State University of West Paraná, Francisco Beltrão - Paraná 85601-970, Brazil
| | - Stephany Christiane Correa
- Center for Bone Marrow Transplantation, Laboratory of Stem Cells, National Cancer Institute (INCA), Rio de Janeiro 20230-130, RJ, Brazil
| | - Thalita Basso Scandolara
- Laboratory of Tumor Biology, State University of West Paraná, Francisco Beltrão - Paraná 85601-970, Brazil.,Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, RJ, Brazil
| | - Janaína Carla da Silva
- Laboratory of Tumor Biology, State University of West Paraná, Francisco Beltrão - Paraná 85601-970, Brazil.,Post-Graduation Program in Health-Applied Sciences, State University of West Paraná, Francisco Beltrão - Paraná 85601-970, Brazil
| | - Bruno Ricardo Pires
- Instituto Nacional de Câncer José Alencar Gomes da Silva, Rio de Janeiro 20230-130, RJ, Brazil.,Department of Cellular & Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Carolina Panis
- Laboratory of Tumor Biology, State University of West Paraná, Francisco Beltrão - Paraná 85601-970, Brazil.,Post-Graduation Program in Health-Applied Sciences, State University of West Paraná, Francisco Beltrão - Paraná 85601-970, Brazil
| |
Collapse
|