1
|
Geng S, Shen Y, Zhang C, Wang N, Gao X, Luo X, Shi N. Correlation between UGT1A1 polymorphism and efficacy and toxicity of irinotecan in Chinese cancer patients. Front Pharmacol 2025; 16:1563566. [PMID: 40170723 PMCID: PMC11958982 DOI: 10.3389/fphar.2025.1563566] [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: 01/21/2025] [Accepted: 02/26/2025] [Indexed: 04/03/2025] Open
Abstract
Objective To assess the association between UGT1A1*6/*28 polymorphisms and Irinotecan (IRI) efficacy/toxicity in Chinese cancer patients. Method We systematically searched PubMed, Cochrane, CNKI, and Wanfang databases. Two investigators independently conducted literature screening, data extraction, and meta-analysis using Revman 5.4. Results This study included 19 clinical trials or case-control studies, with a total of 1,698 patients. Meta-analysis showed that, ① There was no correlation between UGT1A1*6 or UGT1A1*28 gene polymorphism and IRI efficacy; ② UGT1A1*6 or UGT1A1*28 gene polymorphisms are associated with grade 3-4 diarrhea, grade 3-4 neutropenia, and grade 3-4 leukopenia, and the above-mentioned toxic reactions are more common in wild types (GG and TA6/6). ③ There was no correlation between UGT1A1*6 and UGT1A1*28 mutations and the efficacy of IRI; ④ The double wild type was more prone to grade 0-2 neutropenia, the single-site variant was more prone to grade 0-2 diarrhea, and the double-site variant was more prone to grade 3-4 neutropenia, but none of them were related to leukopenia. Conclusion UGT1A1*6/*28 polymorphisms predict IRI-induced toxicity severity but not therapeutic efficacy in Chinese patients. These variants may serve as predictive biomarkers for personalized IRI chemotherapy.
Collapse
Affiliation(s)
- Shuai Geng
- Department of Pharmacy, The Ninth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yulong Shen
- Department of Radiotherapy, The Ninth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Chen Zhang
- Department of Medical Imaging, The Ninth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Nan Wang
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Xinyue Gao
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Xinyu Luo
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Ning Shi
- Department of Pharmacy, The Ninth Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
2
|
Zhang H, Felthaus O, Eigenberger A, Klein S, Prantl L. Treg Cell Therapeutic Strategies for Breast Cancer: Holistic to Local Aspects. Cells 2024; 13:1526. [PMID: 39329710 PMCID: PMC11429654 DOI: 10.3390/cells13181526] [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: 07/30/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024] Open
Abstract
Regulatory T cells (Tregs) play a key role in maintaining immune homeostasis and preventing autoimmunity through their immunosuppressive function. There have been numerous reports confirming that high levels of Tregs in the tumor microenvironment (TME) are associated with a poor prognosis, highlighting their role in promoting an immunosuppressive environment. In breast cancer (BC), Tregs interact with cancer cells, ultimately leading to the suppression of immune surveillance and promoting tumor progression. This review discusses the dual role of Tregs in breast cancer, and explores the controversies and therapeutic potential associated with targeting these cells. Researchers are investigating various strategies to deplete or inhibit Tregs, such as immune checkpoint inhibitors, cytokine antagonists, and metabolic inhibition. However, the heterogeneity of Tregs and the variable precision of treatments pose significant challenges. Understanding the functional diversity of Tregs and the latest advances in targeted therapies is critical for the development of effective therapies. This review highlights the latest approaches to Tregs for BC treatment that both attenuate Treg-mediated immunosuppression in tumors and maintain immune tolerance, and advocates precise combination therapy strategies to optimize breast cancer outcomes.
Collapse
Affiliation(s)
- Hanwen Zhang
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz-Josef-Strauss Allee 11, 93053 Regensburg, Germany (L.P.)
| | | | | | | | | |
Collapse
|
3
|
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
|
4
|
Zhang W, He Y, Tang Y, Dai W, Si Y, Mao F, Xu J, Yu C, Sun X. A meta-analysis of application of PD-1/PD-L1 inhibitor-based immunotherapy in unresectable locally advanced triple-negative breast cancer. Immunotherapy 2023; 15:1073-1088. [PMID: 37337734 DOI: 10.2217/imt-2023-0023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2023] Open
Abstract
Aims: The purpose of this study was to explore the efficacy of immunotherapy for patients with triple-negative breast cancer (TNBC). Materials & methods: Randomized clinical trials comparing immunotherapy with chemotherapy for advanced TNBC patients were included. Results: A total of six articles (3183 patients) were eligible for this meta-analysis. PD-1/PD-L1 inhibitor-based immunotherapy combined with chemotherapy can significantly increase the progression-free survival (hazard ratio [HR] = 0.82; 95% CI = 0.76-1.14; p < 0.001) of unresectable locally advanced or metastatic TNBC patients without effect on overall survival, compared with chemotherapy. Conclusion: PD-1/PD-L1 inhibitors-based immunotherapy can safely improve progression-free survival in patients with unresectable locally advanced or metastatic TNBC, but has no effect on overall survival.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Breast Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, 315010, China
| | - Yujing He
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Yuning Tang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Wei Dai
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Yuexiu Si
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Feiyan Mao
- Department of General Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, 315010, China
| | - Jiaxuan Xu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Chiyuan Yu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Xing Sun
- Department of General Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, 315010, China
| |
Collapse
|
5
|
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: 11] [Impact Index Per Article: 3.7] [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
|
6
|
PD-L1 Expression in Triple-negative Breast Cancer-a Comparative Study of 3 Different Antibodies. Appl Immunohistochem Mol Morphol 2022; 30:726-730. [PMID: 36165931 PMCID: PMC9983741 DOI: 10.1097/pai.0000000000001062] [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: 11/29/2021] [Accepted: 08/12/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Assessment of programmed death protein-ligand 1 (PD-L1) in triple-negative breast cancer (TNBC) has entered daily practice to identify patients eligible for treatment with immune checkpoint inhibitors. However, different antibodies and different cut-offs for PD-L1 positivity are used, and the interchangeability of these methods is not clear. The aim of our study was to analyze whether different PD-L1 antibodies can be used interchangeably to identify TNBC patients as PD-L1 positive. METHODS A tissue microarray encompassing 147 TNBC cases was immunohistochemically analyzed using 3 different antibodies against PD-L1: SP142, SP263, and E1L3N. PD-L1 positivity was determined as ≥1% of positive tumor-associated immune cells. The staining patterns of the 3 antibodies were compared and correlated with clinicopathological data. RESULTS A total of 84 cases were evaluable for PD-L1 analysis with all 3 antibodies. PD-L1 was positive in 50/84 patients (59.5%) with SP263, in 44/84 (52.4%) with E1L3N, and in 29/84 (34.5%) with SP142. There was no statistical difference between the performance of SP263 and E1L3N, but both antibodies stained significantly more cases than the SP142 antibody. CONCLUSIONS Our results show that the 3 PD-L1 antibodies identify different TNBC patient subgroups as PD-L1 positive and, therefore cannot be used interchangeably. Additional studies are needed to further investigate the use and impact of different PD-L1 antibody clones for predictive selection of TNBC patients for treatment with immune checkpoint inhibitors.
Collapse
|
7
|
Zhou Y, Zhao S, Wu T, Zhang H. Comparison of Adverse Reactions Caused by Olaparib for Different Indications. Front Pharmacol 2022; 13:968163. [PMID: 35910367 PMCID: PMC9326361 DOI: 10.3389/fphar.2022.968163] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 06/24/2022] [Indexed: 11/13/2022] Open
Abstract
Objective: Meta-analysis of safety of Olaparib in the treatment of different indications. Methods: The databases of PubMed, The Cochrane Library, EMbase, CNKI, WanFang Data and VIP were searched by computer to collect the research on the indications and the incidence of adverse reactions caused by Olaparib for different cancer types. The search time was from the establishment of the database to May 2022. After two researchers independently screened the literature, extracted the data and evaluated the bias risk included in the study, we used RevMan 5.4 software for meta-analysis. Results: A total of 14 studies were included, with a total sample size of 5119 cases. By meta-analysis, the adverse reactions of Olaparib in the treatment of pancreatic cancer, breast cancer and ovarian cancer were compared. In adverse reactions of any grade, the results showed that fatigue (RR = 1.58, 95% CI [1.20–2.07], p = 0.001) was the most serious in the treatment of pancreatic cancer with Olaparib. Anemia (RR = 2.94, 95% CI [1.97–4.39], p < 0.00001), neutropenia (RR = 1.37, 95% CI [0.80–2.33], p = 0.25), nausea (RR = 1.93, 95% CI [1.61–2.32], p < 0.00001) and vomiting (RR = 1.96, 95% CI [1.59–2.41], p < 0.00001) were the most severe in ovarian cancer. In adverse reactions of grade 3 or above, fatigue (RR = 3.44, 95% CI [1.48–7.98], p = 0.004) and vomiting (RR = 1.09, 95% CI [0.42–2.81], p = 0.86) were the most serious adverse reactions in the treatment of breast cancer with Olaparib. Anemia (RR = 9.74, 95% CI [2.75–34.47], p = 0.0004), neutropenia (RR = 1.33, 95% CI [0.87–2.02], p = 0.19) and nausea (RR = 2.94, 95% CI [1.18–7.32], p = 0.02) were the most severe in ovarian cancer. In addition, the incidence of decreased white blood cell count and hepatotoxicity in the treatment of breast cancer, and the incidence of decreased platelet count, constipation and abdominal pain in the treatment of ovarian cancer were higher than those in pancreatic cancer. Conclusion: Current evidence showed that the risk of adverse reactions of Olaparib in the treatment of different indications is different, and specific analysis and treatment should be carried out for different cancer types. Due to the limitation of the quantity and quality of the included studies, the above conclusions need to be verified by more high-quality studies.
Collapse
Affiliation(s)
- Yujing Zhou
- Department of Nuclear Medicine, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shengwen Zhao
- Department of Radiology, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| | - Tong Wu
- Department of Radiology, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| | - Han Zhang
- Department of Nuclear Medicine, Harbin Medical University Cancer Hospital, Harbin, China
- *Correspondence: Han Zhang,
| |
Collapse
|
8
|
Doney K, Leisenring W, Linden H. Allogeneic hematopoietic cell transplantation in patients with a hematologic malignancy and a prior history of breast cancer. Breast Cancer Res Treat 2022; 194:507-516. [PMID: 35779160 DOI: 10.1007/s10549-022-06658-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 06/08/2022] [Indexed: 11/02/2022]
Abstract
PURPOSE To compare the outcome of allogeneic stem cell transplantation for myeloid malignancies in breast cancer survivors to a contemporaneous control group. METHODS Medical records of all patients with a history of breast cancer who received allogeneic stem cell transplants at a single, tertiary referral Comprehensive Cancer Center between 2002 and 2019 were reviewed. Transplant outcomes were compared to 289 control patients without a history of breast cancer from the same time period. Main outcomes included survival, disease-free survival, non-relapse mortality, relapse or progression of hematologic malignancy, and incidence of recurrent breast cancer after hematopoietic cell transplantation. Comparisons between women with a history of breast cancer and controls utilized propensity score weighting to balance patient characteristics. RESULTS Forty women, ages 30-74 years, with a history of breast cancer received an allogeneic hematopoietic cell transplant for a hematologic malignancy between December 2002 and February 2019. Twelve of the 40 patients are alive with a median survival of 7.4 years (range, 1.9-16.8 years). None of the patients had evidence of recurrent breast cancer prior to death or date of last contact. In multivariable Cox models, all transplant outcomes were similar between the patients and the control group with hematopoietic cell transplant comorbidity score as the most important confounding factor for adjustment in these models. CONCLUSION A history of treated breast cancer should not exclude patients from consideration for allogeneic hematopoietic cell transplantation.
Collapse
Affiliation(s)
- Kristine Doney
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N, D5-280, PO Box 19024, Seattle, WA, 98109-1024, USA. .,University of Washington Medical Center, Seattle, WA, USA.
| | - Wendy Leisenring
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N, D5-280, PO Box 19024, Seattle, WA, 98109-1024, USA.,University of Washington Medical Center, Seattle, WA, USA
| | - Hannah Linden
- University of Washington Medical Center, Seattle, WA, USA.,Seattle Cancer Care Alliance, Seattle, WA, USA
| |
Collapse
|
9
|
Chen Q, Zhang Z, Li X, Bu L. Chemotherapy Combined With Immunotherapy as a First-Line Treatment Brings Benefits to Patients With Lung Squamous Cell Carcinoma but Different Risks of Adverse Reactions: A Systematic Review and Meta-Analysis. Front Pharmacol 2022; 13:940567. [PMID: 35847044 PMCID: PMC9283920 DOI: 10.3389/fphar.2022.940567] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 05/26/2022] [Indexed: 11/30/2022] Open
Abstract
Objective To explore the efficacy and safety of chemotherapy combined with immunotherapy as the first-line treatment of advanced or metastatic squamous NSCLC. Methods Two researchers independently searched PubMed, the Cochrane Library, EMBASE, CNKI, Wanfang Data, and other databases by using a computer, collected the clinical trials or randomized controlled trials published by April 2022 about immunotherapy combined with chemotherapy as the first-line treatment of advanced or metastatic squamous NSCLC, screened the literature, and extracted the data according to the nanodischarge criteria. We used Revman5.4 for statistical analysis of the included studies, and publication bias was analyzed with Egger’s test in Stata12. Results A total of seven clinical trials were included, including 1,510 cases in the chemotherapy combined with the immunotherapy group and 1,519 cases in the chemotherapy group. In terms of effectiveness, compared with the chemotherapy group, chemotherapy combined with immunotherapy for advanced or metastatic squamous NSCLC had longer overall survival (HR = 1.59, 95% CI: 1.46–1.72, p < 0.00001) and progression-free survival (HR = 1.84, 95% CI: 1.66–2.03, p < 0.00001). In terms of safety, the chemotherapy combined with immunotherapy group has a higher risk of adverse reactions at any level and above three levels of hematotoxicity, gastrointestinal abnormalities, and liver dysfunction than the chemotherapy group. Egger’s test has minor publication bias. Conclusion Chemotherapy combined with immunotherapy is effective as the first-line treatment for advanced or metastatic squamous NSCLC, but the risk of adverse reactions is relatively high. If there are adverse reactions in clinical application, it should be treated in time.
Collapse
Affiliation(s)
- Qian Chen
- Department of Pharmacy, Beijing Gaobo Boren Hospital, Beijing, China
| | - Zhen Zhang
- Department of Pharmacy, Beijing Gaobo Boren Hospital, Beijing, China
| | - Xiaoli Li
- Department of Pharmacy, Beijing Gaobo Boren Hospital, Beijing, China
| | - Lingbiao Bu
- Department of Anesthesiology, Zoucheng People’s Hospital, Jining, China
- *Correspondence: Lingbiao Bu,
| |
Collapse
|
10
|
Tang Y, Wang S, Li Y, Yuan C, Zhang J, Xu Z, Hu Y, Shi H, Wang S. Simultaneous glutamine metabolism and PD-L1 inhibition to enhance suppression of triple-negative breast cancer. J Nanobiotechnology 2022; 20:216. [PMID: 35524267 PMCID: PMC9074360 DOI: 10.1186/s12951-022-01424-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 04/18/2022] [Indexed: 11/24/2022] Open
Abstract
Blockade of programmed cell death 1 ligand (PD-L1) has been used to treat triple-negative breast cancer (TNBC), and various strategies are under investigation to improve the treatment response rate. Inhibition of glutamine metabolism can reduce the massive consumption of glutamine by tumor cells and meet the demand for glutamine by lymphocytes in tumors, thereby improving the anti-tumor effect on the PD-L1 blockade therapy. Here, molybdenum disulfide (MoS2) was employed to simultaneously deliver anti-PDL1 antibody (aPDL1) and V9302 to boost the anti-tumor immune response in TNBC cells. The characterization results show that MoS2 has a dispersed lamellar structure with a size of about 181 nm and a size of 232 nm after poly (L-lysine) (PLL) modification, with high stability and biocompatibility. The loading capacity of aPDL1 and V9302 are 3.84% and 24.76%, respectively. V9302 loaded MoS2 (MoS2-V9302) can effectively kill 4T1 cells and significantly reduce glutamine uptake of tumor cells. It slightly increases CD8+ cells in the tumor and promotes CD8+ cells from the tumor edge into the tumor core. In vivo studies demonstrate that the combination of aPDL1 and V9302 (MoS2-aPDL1-V9302) can strongly inhibit the growth of TNBC 4T1 tumors. Interestingly, after the treatment of MoS2-aPDL1-V9302, glutamine levels in tumor interstitial fluid increased. Subsequently, subtypes of cytotoxic T cells (CD8+) in the tumors were analyzed according to two markers of T cell activation, CD69, and CD25, and the results reveal a marked increase in the proportion of activated T cells. The levels of cytokines in the corresponding tumor interstitial fluid are also significantly increased. Additionally, during the treatment, the body weights of the mice remain stable, the main indicators of liver and kidney function in the blood do not increase significantly, and there are no obvious lesions in the main organs, indicating low systemic toxicity. In conclusion, our study provides new insights into glutamine metabolism in the tumor microenvironment affects immune checkpoint blockade therapy in TNBC, and highlights the potential clinical implications of combining glutamine metabolism inhibition with immune checkpoint blockade in the treatment of TNBC.
Collapse
Affiliation(s)
- Yuxia Tang
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Siqi Wang
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yang Li
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chen Yuan
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jie Zhang
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ziqing Xu
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yongzhi Hu
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Haibin Shi
- Department of Interventional Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Shouju Wang
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
11
|
Liu S, Geng S, Shi N, Zhang L, Xue W, Li Y, Jiang K. Survival Prediction of Patients Treated With Immune Checkpoint Inhibitors via KRAS/TP53/EGFR-Single Gene Mutation. Front Pharmacol 2022; 13:878540. [PMID: 35401171 PMCID: PMC8984186 DOI: 10.3389/fphar.2022.878540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 03/08/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Immune checkpoint inhibitors (ICIs) have become an effective treatment option for cancer. KRAS, EGFR and TP53 are common mutated oncogenes in cancer whose single gene status may predict the therapeutic effect of clinical ICIs. In this efficacy evaluation, we aimed to clarify whether the single gene mutation status of KRAS, EGFR or TP53 affects the survival benefits of ICIs in cancer patients. Methods: We used PubMed, Cochrane Library, web of science, and clinical trials Gov database to retrieve qualified documents, the time was up to January 2022. Hazard ratios (HRS) and 95% confidence intervals (CIs) were used to determine the single gene mutation status and no progression of KRAS, EGFR or TP53. Results: A total of 19 studies included 7029 cancer patients treated with ICIs. The results showed that KRAS, EGFR or TP53 single gene mutation could significantly improve PFS and OS in patients receiving ICIs, but the degree of improvement was different. The risk of prolongation of PFS (HR = 1.48, 95% CI = 1.19-1.85, p = 0.0004) and OS (HR = 1.68, 95% CI = 1.36-2.07, p < 0.00001) caused by TP53 single gene mutation was relatively high, the risk ratio of prolongation of PFS (HR = 1.38, 95% CI = 1.21-1.57, p < 0.00001) and OS (HR = 1.56, 95% CI = 1.20-2.04, p = 0.001) caused by EGFR single gene mutation was the second, the risk ratio of prolongation of PFS (HR = 1.33, 95% CI = 1.12-1.57, p = 0.001) and OS (HR = 1.39, 95% CI = 1.18-1.63, p < 0.00001) caused by KRAS single gene mutation was relatively low, and the results were significantly different. Conclusion: In cancer patients, KRAS, EGFR or TP53 single gene status is correlated with the benefits of immunotherapy PFS and OS, which suggests that gene sequencing should be carried out in time in the process of clinical treatment to determine the gene mutation of patients and better predict the clinical treatment effect of ICIs.
Collapse
Affiliation(s)
- Shui Liu
- Department of Pharmacy, Emergency General Hospital, Beijing, China
| | - Shuai Geng
- Department of Pharmacy, Strategic Support Force Medical Center, Beijing, China
| | - Ning Shi
- Department of Pharmacy, Strategic Support Force Medical Center, Beijing, China
| | - Lili Zhang
- Department of Pharmacy, Emergency General Hospital, Beijing, China
| | - Wenxin Xue
- Department of Pharmacy, Emergency General Hospital, Beijing, China
| | - Yiwen Li
- Department of Pharmacy, Emergency General Hospital, Beijing, China
| | - Kai Jiang
- Department of Gastroenterology, Traditional Chinese Medical Hospital of Zhuji, Zhuji, China
| |
Collapse
|
12
|
Chen Q, Li X, Zhang Z, Wu T. Systematic Review of Olaparib in the Treatment of Recurrent Platinum Sensitive Ovarian Cancer. Front Oncol 2022; 12:858826. [PMID: 35299755 PMCID: PMC8921481 DOI: 10.3389/fonc.2022.858826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 02/08/2022] [Indexed: 11/13/2022] Open
Abstract
Objective To systematically evaluate the efficacy and safety of olaparib in the treatment of recurrent platinum-sensitive ovarian cancer. Methods The Cochrane Library, PubMed, Chinese Biomedical Literature Database, CNKI, VIP Database, Wanfang Science and Technology Database were searched for randomized controlled trials (RCTs) of olaparib in the treatment of recurrent platinum-sensitive ovarian cancer from the establishment of each database to January 2022. Two reviewers independently evaluated the quality of the literature, extracted the data, and cross-checked the methodological quality. Meta-analysis was performed using RevMan 5.4 software. Results A total of 7 RCTs were included, including 2406 patients, There were 1497 patients in treatment groups and 909 patients in the control group. Meta-analysis results showed that in terms of effectiveness, the overall survival time of patients in the olaparib group [HR=1.24, 95%CI(1.06, 1.45), P=0.006]; in terms of safety, for all grades of adverse events (including nausea, fatigue, vomiting, diarrhea, abdominal pain, and headache), [HR=1.54, 95%CI(1.38, 1.71), P=0.0002], for grade 3 or higher adverse events (including nausea, fatigue, vomiting, diarrhea, abdominal pain, and headache), [HR=2.13, 95%CI(1.61, 2.81), P=0.003], there were significant differences compared with the control group, suggesting that the risk of adverse reactions in the experimental group was higher than that in the control group. Subgroup analysis showed that only abdominal pain, headache and vomiting were not statistically significant, and other adverse reactions were statistically significant. Conclusion Based on the existing clinical evidence, olaparib in the treatment of recurrent platinum-sensitive ovarian cancer has a longer overall survival than the control group. It is an ideal regimen, but the incidence of adverse reactions is high.
Collapse
Affiliation(s)
- Qian Chen
- Department of Pharmacy, Beijing Gaobo Boren Hospital, Beijing, China
| | - Xiaoli Li
- Department of Pharmacy, Beijing Gaobo Boren Hospital, Beijing, China
| | - Zhen Zhang
- Department of Pharmacy, Beijing Gaobo Boren Hospital, Beijing, China
| | - Tong Wu
- Department of Pharmacy, Beijing Gaobo Boren Hospital, Beijing, China
| |
Collapse
|
13
|
Jiang S, Geng S, Chen Q, Zhang C, Cheng M, Yu Y, Zhang S, Shi N, Dong M. Effects of Concomitant Antibiotics Use on Immune Checkpoint Inhibitor Efficacy in Cancer Patients. Front Oncol 2022; 12:823705. [PMID: 35223505 PMCID: PMC8864310 DOI: 10.3389/fonc.2022.823705] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 01/14/2022] [Indexed: 12/30/2022] Open
Abstract
Objective Immune checkpoint inhibitors (ICIs) have changed the outcomes of a variety of cancers in an unprecedented manner. Gut microbiome plays a crucial regulatory role in the antineoplastic therapy of ICIs, which can be influenced by antibiotic (ABX) administration. In this efficacy evaluation, we aimed to clarify the correlations of ABX administration with the survival of cancer patients receiving ICIs treatment. Method The eligible literatures were searched using PubMed, Cochrane Library, Web of Science, and Clinical trials.gov databases before Nov 2021. The correlations of ABX administration with progression-free survival (PFS) and overall survival (OS) were determined using Hazard ratios (HRs) coupled with 95% confidence intervals (CIs). Results A total of 12 studies enrolling 6010 cancer patients receiving ICIs treatment were included in this efficacy evaluation. ABX administration was significantly correlated worse PFS (HR=1.60, 95%CI=1.33-1.92, P<0.00001) and OS (HR=1.46, 95%CI=1.32-1.61, P<0.00001). Similar results were found in the subgroup analysis of non-small cell lung cancer (NSCLC), renal cell carcinoma (RCC) and melanoma. Conclusions ABX use during ICIs treatment of cancer may significantly shorten PFS and OS. ABX should be used cautiously in cancer patients receiving ICIs. However, further validations are still essential due to existing publication bias.
Collapse
Affiliation(s)
- Shuai Jiang
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shuai Geng
- Department of Pharmacy, Strategic Support Force Medical Center, Beijing, China
| | - Qian Chen
- Department of Pharmacy, Beijing Boren Hospital, Beijing, China
| | - Chen Zhang
- Department of Medical Imaging, Strategic Support Force Medical Center, Beijing, China
| | - Mengfei Cheng
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yang Yu
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shuo Zhang
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ning Shi
- Department of Pharmacy, Strategic Support Force Medical Center, Beijing, China
| | - Mei Dong
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
14
|
Zhu L, Liu J, Chen J, Zhou Q. The developing landscape of combinatorial therapies of immune checkpoint blockade with DNA damage repair inhibitors for the treatment of breast and ovarian cancers. J Hematol Oncol 2021; 14:206. [PMID: 34930377 PMCID: PMC8686226 DOI: 10.1186/s13045-021-01218-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 12/05/2021] [Indexed: 02/07/2023] Open
Abstract
The use of immune checkpoint blockade (ICB) using antibodies against programmed death receptor (PD)-1, PD ligand (PD-L)-1, and cytotoxic T-lymphocyte antigen 4 (CTLA-4) has redefined the therapeutic landscape in solid tumors, including skin, lung, bladder, liver, renal, and breast tumors. However, overall response rates to ICB therapy remain limited in PD-L1-negative patients. Thus, rational and effective combination therapies will be needed to address ICB treatment resistance in these patients, as well as in PD-L1-positive patients who have progressed under ICB treatment. DNA damage repair inhibitors (DDRis) may activate T-cell responses and trigger inflammatory cytokines release and eventually immunogenic cancer cell death by amplifying DNA damage and generating immunogenic neoantigens, especially in DDR-defective tumors. DDRi may also lead to adaptive PD-L1 upregulation, providing a rationale for PD-L1/PD-1 blockade. Thus, based on preclinical evidence of efficacy and no significant overlapping toxicity, some ICB/DDRi combinations have rapidly progressed to clinical testing in breast and ovarian cancers. Here, we summarize the available clinical data on the combination of ICB with DDRi agents for treating breast and ovarian cancers and discuss the mechanisms of action and other lessons learned from translational studies conducted to date. We also review potential biomarkers to select patients most likely to respond to ICB/DDRi combination therapy.
Collapse
Affiliation(s)
- Lingling Zhu
- Lung Cancer Center, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Jiewei Liu
- Lung Cancer Center, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Jiang Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, Zhejiang Province, China.
| | - Qinghua Zhou
- Lung Cancer Center, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan Province, China.
| |
Collapse
|
15
|
Palma MB, Tronik-Le Roux D, Amín G, Castañeda S, Möbbs AM, Scarafia MA, La Greca A, Daouya M, Poras I, Inda AM, Moro LN, Carosella ED, García MN, Miriuka SG. HLA-G gene editing in tumor cell lines as a novel alternative in cancer immunotherapy. Sci Rep 2021; 11:22158. [PMID: 34773056 PMCID: PMC8589947 DOI: 10.1038/s41598-021-01572-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 10/22/2021] [Indexed: 11/08/2022] Open
Abstract
Cancer immunotherapies based mainly on the blockade of immune-checkpoint (IC) molecules by anti-IC antibodies offer new alternatives for treatment in oncological diseases. However, a considerable proportion of patients remain unresponsive to them. Hence, the development of novel clinical immunotherapeutic approaches and/or targets are crucial.W In this context, targeting the immune-checkpoint HLA-G/ILT2/ILT4 has caused great interest since it is abnormally expressed in several malignancies generating a tolerogenic microenvironment. Here, we used CRISPR/Cas9 gene editing to block the HLA-G expression in two tumor cell lines expressing HLA-G, including a renal cell carcinoma (RCC7) and a choriocarcinoma (JEG-3). Different sgRNA/Cas9 plasmids targeting HLA-G exon 1 and 2 were transfected in both cell lines. Downregulation of HLA-G was reached to different degrees, including complete silencing. Most importantly, HLA-G - cells triggered a higher in vitro response of immune cells with respect to HLA-G + wild type cells. Altogether, we demonstrated for the first time the HLA-G downregulation through gene editing. We propose this approach as a first step to develop novel clinical immunotherapeutic approaches in cancer.
Collapse
Affiliation(s)
- María Belén Palma
- Cátedra de Citología, Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
- LIAN-CONICET, Fundación FLENI, Buenos Aires, Argentina
| | - Diana Tronik-Le Roux
- Atomic Energy and Alternative Energies Agency (CEA), Hematology and Immunology Research Division, Saint-Louis Hospital, Paris, France
- U976 HIPI Unit, University of Paris, Paris, France
| | | | | | - Alan M Möbbs
- LIAN-CONICET, Fundación FLENI, Buenos Aires, Argentina
| | | | | | - Marina Daouya
- Atomic Energy and Alternative Energies Agency (CEA), Hematology and Immunology Research Division, Saint-Louis Hospital, Paris, France
- U976 HIPI Unit, University of Paris, Paris, France
| | - Isabelle Poras
- Atomic Energy and Alternative Energies Agency (CEA), Hematology and Immunology Research Division, Saint-Louis Hospital, Paris, France
- U976 HIPI Unit, University of Paris, Paris, France
| | - Ana María Inda
- Cátedra de Citología, Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
- Comisión de Investigaciones Científicas (CIC), Buenos Aires, Argentina
| | - Lucía N Moro
- LIAN-CONICET, Fundación FLENI, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Edgardo D Carosella
- Atomic Energy and Alternative Energies Agency (CEA), Hematology and Immunology Research Division, Saint-Louis Hospital, Paris, France
- U976 HIPI Unit, University of Paris, Paris, France
| | - Marcela N García
- Cátedra de Citología, Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina.
| | - Santiago G Miriuka
- LIAN-CONICET, Fundación FLENI, Buenos Aires, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
16
|
Lazarus G, Budiman RA, Rinaldi I. Does immune checkpoint inhibitor increase the risks of poor outcomes in COVID-19-infected cancer patients? A systematic review and meta-analysis. Cancer Immunol Immunother 2021; 71:373-386. [PMID: 34173850 PMCID: PMC8233621 DOI: 10.1007/s00262-021-02990-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 06/16/2021] [Indexed: 12/17/2022]
Abstract
Background The association between immune checkpoint inhibitor (ICI) and outcomes of cancer patients with coronavirus disease 2019 (COVID-19) infection has yet to be systematically evaluated. This meta-analysis aims to investigate the effects of ICI treatment on COVID-19 prognosis, including mortality, severity, and any other prognosis-related outcomes. Methods Eligible studies published up to 27 February 2021 were included and assessed for risk of bias using the Quality in Prognosis Studies tool. A random-effects meta-analysis was conducted to estimate the pooled effect size along with its 95% confidence intervals. The quality of body evidence was evaluated using the modified Grading of Recommendations Assessment, Development, and Evaluation framework. Results Eleven studies involving a total of 2826 COVID-19-infected cancer patients were included in the systematic review. We discovered a moderate-to-high quality of evidence that ICI was not associated with a higher mortality risk, while the other outcomes yielded a very low-to-low-evidence quality. Although our findings indicated that ICI did not result in a higher risk of severity and hospitalization, further evidence is required to confirm our findings. In addition, we discovered that prior exposure to chemoimmunotherapy may be linked with a higher risk of COVID-19 severity (OR 8.19 [95% CI: 2.67–25.08]; I2 = 0%), albeit with small sample size. Conclusion Our findings indicated that ICI treatment should not be adjourned nor terminated during the current pandemic. Rather, COVID-19 vigilance should be increased in such patients. Further studies with larger cohorts and higher quality of evidence are required to substantiate our findings. Trial registration number This project has been prospectively registered at PROSPERO (registration ID: CRD42020202142) on 4 August 2020. Supplementary Information The online version contains supplementary material available at 10.1007/s00262-021-02990-9.
Collapse
Affiliation(s)
- Gilbert Lazarus
- Faculty of Medicine, Universitas Indonesia, Jl. Salemba Raya No. 6, RW 5, Kenari, Kec. Senen, Kota Jakarta Pusat, Jakarta, 10430, Indonesia.
| | - Refael Alfa Budiman
- Faculty of Medicine, Universitas Indonesia, Jl. Salemba Raya No. 6, RW 5, Kenari, Kec. Senen, Kota Jakarta Pusat, Jakarta, 10430, Indonesia
| | - Ikhwan Rinaldi
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia - Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| |
Collapse
|
17
|
Ali MA, Aiman W, Shah SS, Hussain M, Kashyap R. Efficacy and safety of pembrolizumab based therapies in triple-negative breast cancer: A systematic review of clinical trials. Crit Rev Oncol Hematol 2020; 157:103197. [PMID: 33309890 DOI: 10.1016/j.critrevonc.2020.103197] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/02/2020] [Accepted: 12/06/2020] [Indexed: 02/08/2023] Open
Abstract
Breast cancer is the most common cause of cancer-related deaths among women. There are a limited number of targeted therapies available for triple-negative breast cancer (TNBC), and chemotherapy is the mainstay of treatment. Among checkpoint inhibitors, atezolizumab is the only drug approved for PD-L1+ TNBC patients. We performed a systematic review to assess the efficacy and safety of PD-1 inhibitor pembrolizumab in triple-negative breast cancer. We included 15 clinical trials in this review. Pembrolizumab was well tolerated by all patients with triple-negative breast cancer. Pembrolizumab was more effective in the treatment of early-stage TNBC patients as compared to placebo, regardless of PD-L1 status. In advanced-stage breast cancer, pembrolizumab was as effective as single-agent chemotherapy with a better safety profile. Pembrolizumab with chemotherapy showed significantly better median progression free survival as compared to chemotherapy in advanced TNBC.
Collapse
Affiliation(s)
| | | | - Syed S Shah
- Department of Hospital Medicine, University of Kentucky, Lexington, USA.
| | | | | |
Collapse
|