1
|
Alwhaibi A, Alenazi MA, Alghadeer S, Mansy W, Alsaif RA, Abualreesh NE, Alanazi RJ, Alroumi A, Alanazi SA. A Real-World Comparison of the Safety Profile for Immune Checkpoint Inhibitors in Oncology Patients. J Clin Med 2025; 14:388. [PMID: 39860394 PMCID: PMC11765622 DOI: 10.3390/jcm14020388] [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: 12/11/2024] [Revised: 01/02/2025] [Accepted: 01/05/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Owing to the growing use of immune checkpoint inhibitors (ICIs) in the treatment of cancer, a wide spectrum of toxicity has arisen among cancer patients. Yet, limited ICI toxicity-related research is currently conducted in our region. Methods: This is a retrospective observational study conducted on adult cancer patients who received at least one cycle of ICI single therapy. Toxicity profiles of different ICI monotherapies were described and compared, and their association with different risk factors was assessed. SPSS version 28 was used for statistical analyses, and p < 0.05 was considered statistically significant. Results: A total of 428 patients were treated with anti-PD1 (nivolumab [n = 221, 51.6%] and pembrolizumab [n = 126, 29.5%]) or anti-PD-L1 (atezolizumab [n = 78, 18.2%] and durvalumab [n = 3, 0.7%]). Pneumonia was the most common complication (10%), followed by acute kidney injury (AKI; 8.2%) and hepatitis (7.9%). The proportion of hepatitis cases was significantly higher among atezolizumab compared to nivolumab-, pembrolizumab-, and durvalumab-treated patients (17.95% vs. 7.7% vs. 2.4% vs. 0.0%, respectively; p < 0.001). Gastrointestinal complication (colitis) was detected in 3.3% of patients with a significant difference between treatment groups (4.5%, 1.6%, 1.3%, and 33.3% in nivolumab, pembrolizumab, atezolizumab, and durvalumab, respectively; p = 0.008). Cardiac complications occurred in 1.2% of patients with a significant difference between treatment groups (0.5% in the nivolumab, 3.8% in the atezolizumab, 33.3% in the durvalumab, and none in the pembrolizumab groups (p < 0.001)). Musculoskeletal side effects, including both arthralgia and fatigue, were the most-reported side effects by 39.5% of patients, with significantly higher arthralgia complainers only in nivolumab (7.7%) compared to other treatment groups (0%, 2.6%, and 0% in pembrolizumab, atezolizumab, and durvalumab, respectively, p = 0.007). Hepatic, cardiovascular, hematological, respiratory, renal, gastrointestinal complications, thyroid complications, and dermatological side effects were found to occur on weeks 6, 7.5, 8, 8, 10, 10, 10.5, and 12 after treatment initiation, respectively, with no significant difference between treatment groups. Despite that, hepatitis and AKI tended to occur earlier with atezolizumab (week 2, p = 0.084) and pembrolizumab (week 2, p = 0.062), respectively, compared to their comparators. The female gender and a history of hepatitis were found to increase the odds of hepatic complication with anti-PD1 or anti-PD-L1 use [OR = 2.71; 95% CI 1.07-6.85, OR = 11.14; 95% CI 3.46-35.88, respectively]. Previous exposure to cancer therapy only was found to increase the odds of developing pneumonia among the treated patients [OR = 3.08; 95% CI 1.12-8.85]. Having hematological malignancy influenced the odds of hematological complications positively (either neutropenia or thrombocytopenia) compared to solid malignancies when patients were treated with anti-PD1 or anti-PD-L1 [OR = 17.18; 95% CI 4.06-72.71]. Finally, the female gender was found to positively associate with the odds of nausea/vomiting and fatigue secondary to anti-PD1 or anti-PD-L1 administration [OR = 2.08; 95% CI 1.34-3.21, OR = 1.65; 95% CI 1.09-2.51, respectively]. On the other hand, previous exposure to cancer therapy was found to reduce the risk of having arthralgia with anti-PD1 or anti-PD-L1 administration [OR = 0.344; 95% CI 0.121-0.974]. Conclusions: Treatment with anti-PD1 or anti-PD-L1 was associated with a spectrum of complications and side effects. Several risk factors have been identified to impact their occurrence. ICI toxicities and risk factors influencing their odds should be recognized and considered in clinical practice, as this could help in individualizing therapeutics regimens and avoiding treatment interruption.
Collapse
Affiliation(s)
- Abdulrahman Alwhaibi
- Department of Clinical Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.A.); (W.M.); (N.E.A.)
| | - Miteb A. Alenazi
- Pharmacy Department, King Saud University Medical City, Riyadh 11411, Saudi Arabia;
| | - Sultan Alghadeer
- Department of Clinical Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.A.); (W.M.); (N.E.A.)
| | - Wael Mansy
- Department of Clinical Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.A.); (W.M.); (N.E.A.)
| | - Reem A. Alsaif
- Pharmacy Department, King Abdulaziz Medical City, National Guard Health Affairs, Riyadh 11426, Saudi Arabia;
| | - Nawaf E. Abualreesh
- Department of Clinical Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.A.); (W.M.); (N.E.A.)
| | - Rakan J. Alanazi
- Pharmacy Practice Department, College of Pharmacy, Alfaisal University, Riyadh 11533, Saudi Arabia;
| | - Abdullah Alroumi
- College of Pharmacy, King Saud Bin Abdulaziz University for Health Sciences, Riyadh 11461, Saudi Arabia; (A.A.); (S.A.A.)
| | - Saleh A. Alanazi
- College of Pharmacy, King Saud Bin Abdulaziz University for Health Sciences, Riyadh 11461, Saudi Arabia; (A.A.); (S.A.A.)
| |
Collapse
|
2
|
Liguori L, Giorgio G, Polcaro G, Pagliara V, Malandrino D, Perri F, Cascella M, Ottaiano A, Conti V, Servetto A, Bianco R, Pepe S, Sabbatino F. Checkpoint based immunotherapy in non-small cell lung cancer: a real-world retrospective study. Front Immunol 2024; 15:1419544. [PMID: 39664396 PMCID: PMC11631946 DOI: 10.3389/fimmu.2024.1419544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 11/06/2024] [Indexed: 12/13/2024] Open
Abstract
Introduction Immune checkpoint inhibitor (ICI)-based immunotherapy targeting programmed cell death 1 (PD-1) or its ligand 1 (PD-L1) has radically changed the management of many types of solid tumors including non-small cell lung cancer (NSCLC). Many clinical trials have demonstrated that ICIs improve the survival and the quality of life of patients with advanced non oncogene NSCLC as compared to standard therapies. However, not all patients achieve a clinical benefit from this immunotherapeutic approach. As a result, real-word validation of the efficacy and safety of ICIs can be useful for defining potential predictive biomarkers as well as for overcoming limitations linked to clinical trial restrictions. Methods We retrospectively retrieved the clinical data of patients with advanced non oncogene NSCLC treated with ICIs (anti-PD-1 or anti-PD-L1) as single agent or in combination with chemotherapy at "San Giovanni di Dio e Ruggi D'Aragona" University Hospital from January 2016 to December 2023. Potential correlations between clinical-pathological characteristics and safety or survival outcomes were investigated employing the Fisher's exact test, Mann-Whitney U test, the Kruskal-Wallis method and log-rank test, as applicable. Multivariate survival analyses were performed using the Cox proportional hazards model. Results Clinical data of 129 patients were retrieved. At a median follow-up of 29.70 months, progression-free survival (PFS) and overall survival (OS) were 5.27 months and 8.43 months, respectively. At the multivariate analyses, smoking status, presence of bone metastases and the occurrence of immune-related adverse events (irAEs) were correlated with both PFS and OS. Moreover, patients treated with anti-PD-1-based therapy achieved an increased clinical benefit than those treated with anti-PD-L1. Discussion In this study we described our real-world experience of ICIs for the treatment of patients with advanced non oncogene NSCLC. A decreased OS in our study population was reported as compared to that of patients included in the clinical trials. Noteworthy, correlations between clinical-pathological characteristics and survival outcomes emerged. Nevertheless, the potential integration of clinical-pathological characteristics as predictive biomarkers in more accurate therapeutic algorithms as well as the underlying biological mechanisms should be further validated in ad hoc studies.
Collapse
Affiliation(s)
- Luigi Liguori
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Gabriele Giorgio
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
| | - Giovanna Polcaro
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
| | - Valentina Pagliara
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
| | - Domenico Malandrino
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
| | - Francesco Perri
- Medical and Experimental Head and Neck Oncology Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy
| | - Marco Cascella
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
| | - Alessandro Ottaiano
- Division of Innovative Therapies for Abdominal Metastases, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy
| | - Valeria Conti
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
| | - Alberto Servetto
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Roberto Bianco
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Stefano Pepe
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
| | - Francesco Sabbatino
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
| |
Collapse
|
3
|
Ben Saad E, Oroya A, Anto NP, Bachais M, Rudd CE. PD-1 endocytosis unleashes the cytolytic potential of checkpoint blockade in tumor immunity. Cell Rep 2024; 43:114907. [PMID: 39471174 DOI: 10.1016/j.celrep.2024.114907] [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: 10/03/2023] [Revised: 09/12/2024] [Accepted: 10/08/2024] [Indexed: 11/01/2024] Open
Abstract
PD-1 immune checkpoint blockade (ICB) is a key cancer treatment. While blocking PD-1 binding to ligand is known, the role of internalization in enhancing ICB efficacy is less explored. Our study reveals that PD-1 internalization helps unlock ICB's full potential in cancer immunotherapy. Anti-PD-1 induces 50%-60% surface PD-1 internalization from human and mouse cells, leaving low to intermediate levels of resistant receptors. Complexes then appear in early and late endosomes. Both CD4 and CD8 T cells, especially CD8+ effectors, are affected. Nivolumab outperforms pembrolizumab in human T cells, while PD-1 internalization requires crosslinking by bivalent antibody. While mono- and bivalent anti-PD-1 inhibit tumor growth with CD8 tumor-infiltrating cells expressing increased granzyme B, bivalent antibody is more effective where the combination of steric blockade and endocytosis induces greater CD8+ T cell tumor infiltration and the expression of the cytolytic pore protein, perforin. Our findings highlight an ICB mechanism that combines steric blockade and PD-1 endocytosis for optimal checkpoint immunotherapy.
Collapse
Affiliation(s)
- Elham Ben Saad
- Department of Medicine, Universite de Montréal, Montréal, QC H3C 3J7, Canada; Centre de Recherche Hopital Maisonneuve-Rosemont, Montréal, Quebec, QC H1T 2M4, Canada; Department of Biochemistry and Molecular Medicine, Universite de Montréal, Montréal, QC H3T 1J4, Canada
| | - Andres Oroya
- Department of Medicine, Universite de Montréal, Montréal, QC H3C 3J7, Canada; Centre de Recherche Hopital Maisonneuve-Rosemont, Montréal, Quebec, QC H1T 2M4, Canada; Department of Microbiology, Infection and Immunology, Universite de Montréal, Montréal, QC, Canada
| | - Nikhil Ponnoor Anto
- Department of Medicine, Universite de Montréal, Montréal, QC H3C 3J7, Canada; Centre de Recherche Hopital Maisonneuve-Rosemont, Montréal, Quebec, QC H1T 2M4, Canada; Department of Microbiology, Infection and Immunology, Universite de Montréal, Montréal, QC, Canada
| | - Meriem Bachais
- Department of Medicine, Universite de Montréal, Montréal, QC H3C 3J7, Canada; Centre de Recherche Hopital Maisonneuve-Rosemont, Montréal, Quebec, QC H1T 2M4, Canada; Department of Microbiology, Infection and Immunology, Universite de Montréal, Montréal, QC, Canada
| | - Christopher E Rudd
- Department of Medicine, Universite de Montréal, Montréal, QC H3C 3J7, Canada; Centre de Recherche Hopital Maisonneuve-Rosemont, Montréal, Quebec, QC H1T 2M4, Canada; Department of Biochemistry and Molecular Medicine, Universite de Montréal, Montréal, QC H3T 1J4, Canada; Department of Microbiology, Infection and Immunology, Universite de Montréal, Montréal, QC, Canada.
| |
Collapse
|
4
|
Miyake M, Nishimura N, Oda Y, Miyamoto T, Iida K, Inoue K, Tachibana A, Yoshikawa T, Sakamoto K, Ohnishi M, Maesaka F, Takamatsu N, Mieda K, Ohmori C, Matsubara T, Tomizawa M, Shimizu T, Ohnishi K, Hori S, Morizawa Y, Gotoh D, Nakai Y, Torimoto K, Tanaka N, Fujimoto K. Difference of oncological efficacy between two immune checkpoint inhibitors following first-line platinum-based chemotherapy in patients with unresectable, metastatic, advanced urothelial carcinoma: a multicenter real-world Japanese cohort. Int J Clin Oncol 2024; 29:1311-1325. [PMID: 38888683 DOI: 10.1007/s10147-024-02573-5] [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: 03/16/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND Maintenance avelumab is currently recommended for patients with unresectable and/or metastatic (mUC) achieving at least stable disease (SD) on first-line platinum-based chemotherapy (1L-CT). Pembrolizumab is an alternative therapeutic avenue for this patient cohort in clinical practice. We investigated real-world data, focusing on the correlation between response to 1L-CT and oncological efficacy of subsequent immune checkpoint inhibitor (ICI) therapy with avelumab or pembrolizumab. METHODS A multicenter database registered 626 patients with mUC diagnosed from 2008-2023; among these, 175 receiving 2-6 cycles of 1L-CT followed by ICI therapy. Patients were categorized based on response to 1L-CT using the Response Evaluation Criteria in Solid Tumors (v1.1). Objective response rate on ICI, progression to ICI-free survival (ICI-PFS), and overall survival from start of 1L-CT were compared between avelumab-treated and pembrolizumab-treated patients in each response subgroup. RESULTS ICI-PFS was significantly longer in patients achieving partial response on 1L-CT and subsequently receiving pembrolizumab compared to those receiving avelumab. Notably, patients achieving SD on 1L-CT and subsequently receiving pembrolizumab manifested significantly higher objective response rate (14% and 41%, respectively) and prolonged ICI-PFS relative to those receiving avelumab. In contrast, overall survival did not delineate difference between patients treated with avelumab versus pembrolizumab. Similar findings were discerned in the subanalysis of patients having favorable SD (tumor shrinkage, from - 29 to 0%) and unfavorable SD (tumor enlargement, from + 1 to + 19%) on 1L-CT. CONCLUSIONS Our study provides real-world evidence regarding difference of oncological efficacy between maintenance avelumab and subsequent pembrolizumab in patients with mUC who achieved partial response or SD on 1L-CT.
Collapse
Affiliation(s)
- Makito Miyake
- Department of Urology, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan.
| | - Nobutaka Nishimura
- Department of Urology, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan
- Department of Urology, Hirao Hospital, Kashihara, Nara, Japan
| | - Yuki Oda
- Department of Urology, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan
| | | | - Kota Iida
- Department of Urology, Tane General Hospital, Osaka, Japan
| | - Kuniaki Inoue
- Department of Urology, Osaka Gyoumeikan Hospital, Osaka, Japan
| | - Akira Tachibana
- Department of Urology, Hoshigaoka Medical Center, Hirakata, Osaka, Japan
| | | | - Keichi Sakamoto
- Department of Urology, Osaka Kaisei Hospital, Yodogawa, Osaka, 532-0003, Japan
| | | | | | - Norimi Takamatsu
- Department of Urology, Yamatotakada Municipal Hospital, Yamatotakada, Nara, Japan
| | - Kosuke Mieda
- Department of Urology, Nara Prefecture General Medical Center, Nara, Japan
| | - Chihiro Ohmori
- Department of Urology, Nara Prefecture General Medical Center, Nara, Japan
| | - Toshihiko Matsubara
- Department of Urology, Matsusaka Chuo General Hospital, Matsusaka, Mie, Japan
| | - Mitsuru Tomizawa
- Department of Urology, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan
| | - Takuto Shimizu
- Department of Urology, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan
| | - Kenta Ohnishi
- Department of Urology, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan
| | - Shunta Hori
- Department of Urology, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan
| | - Yosuke Morizawa
- Department of Urology, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan
| | - Daisuke Gotoh
- Department of Urology, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan
| | - Yasushi Nakai
- Department of Urology, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan
| | - Kazumasa Torimoto
- Department of Urology, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan
| | - Nobumichi Tanaka
- Department of Urology, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan
- Department of Prostate Brachytherapy, Nara Medical University, Kashihara, Nara, 634-8522, Japan
| | - Kiyohide Fujimoto
- Department of Urology, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan
| |
Collapse
|
5
|
Kocikowski M, Dziubek K, Węgrzyn K, Hrabal V, Zavadil-Kokas F, Vojtesek B, Alfaro JA, Hupp T, Parys M. Comparative characterization of two monoclonal antibodies targeting canine PD-1. Front Immunol 2024; 15:1382576. [PMID: 38779661 PMCID: PMC11110041 DOI: 10.3389/fimmu.2024.1382576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 03/11/2024] [Indexed: 05/25/2024] Open
Abstract
Monoclonal antibodies targeting immune checkpoints have revolutionized oncology. Yet, the effectiveness of these treatments varies significantly among patients, and they are associated with unexpected adverse events, including hyperprogression. The murine research model used in drug development fails to recapitulate both the functional human immune system and the population heterogeneity. Hence, a novel model is urgently needed to study the consequences of immune checkpoint blockade. Dogs appear to be uniquely suited for this role. Approximately 1 in 4 companion dogs dies from cancer, yet no antibodies are commercially available for use in veterinary oncology. Here we characterize two novel antibodies that bind canine PD-1 with sub-nanomolar affinity as measured by SPR. Both antibodies block the clinically crucial PD-1/PD-L1 interaction in a competitive ELISA assay. Additionally, the antibodies were tested with a broad range of assays including Western Blot, ELISA, flow cytometry, immunofluorescence and immunohistochemistry. The antibodies appear to bind two distinct epitopes as predicted by molecular modeling and peptide phage display. Our study provides new tools for canine oncology research and a potential veterinary therapeutic.
Collapse
Affiliation(s)
- Mikolaj Kocikowski
- International Centre for Cancer Vaccine Science, University of Gdansk, Gdansk, Poland
- The Royal (Dick) School of Veterinary Studies and The Roslin Institute, University of Edinburgh, Midlothian, United Kingdom
| | - Katarzyna Dziubek
- International Centre for Cancer Vaccine Science, University of Gdansk, Gdansk, Poland
| | - Katarzyna Węgrzyn
- Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, Gdansk, Poland
| | - Vaclav Hrabal
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Filip Zavadil-Kokas
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Borivoj Vojtesek
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Javier Antonio Alfaro
- International Centre for Cancer Vaccine Science, University of Gdansk, Gdansk, Poland
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
- Institute for Adaptive and Neural Computation, School of Informatics, University of Edinburgh, Edinburgh, United Kingdom
| | - Ted Hupp
- International Centre for Cancer Vaccine Science, University of Gdansk, Gdansk, Poland
- Institute of Genetic and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Maciej Parys
- The Royal (Dick) School of Veterinary Studies and The Roslin Institute, University of Edinburgh, Midlothian, United Kingdom
| |
Collapse
|
6
|
Tang YH, Bergmann J, Vaidya D, Faraday N. Association of Preoperative Immune Checkpoint Inhibitor Therapy With Cardiopulmonary Instability and Organ Injury After High-Risk Surgery. Crit Care Explor 2024; 6:e1068. [PMID: 38562380 PMCID: PMC10984666 DOI: 10.1097/cce.0000000000001068] [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] [Indexed: 04/04/2024] Open
Abstract
OBJECTIVES To assess the relationship between prior exposure to immune checkpoint inhibitors (ICIs) and the risk of postoperative complications in cancer patients. DESIGN Single-center retrospective cohort study. INTERVENTIONS The main exposure was treatment with an FDA-approved ICI within 6 months before surgery. MEASUREMENTS AND MAIN RESULTS Exposure to ICIs and covariates was determined from the electronic health record. The primary outcome was a composite of postoperative complications, including prolonged pressor or oxygen dependence, kidney injury, or myocardial injury. Secondary outcomes included each subcomponent of the primary outcome. Of 7674 subjects with cancer admitted to the ICU after surgery, 247 were exposed to one or more ICIs in the 6 months before surgery. After propensity score matching, 197 ICI-exposed subjects were matched to 777 nonexposed. The composite outcome occurred in 70 of 197 (35.5%) ICI-exposed subjects and 251 of 777 (32.3%) nonexposed. There was no difference between exposed and nonexposed groups in the primary composite outcome (odds ratio [OR], 1.12; 95% CI, 0.80-1.58) by conditional logistic regression. Risk of the secondary outcome of prolonged pressor dependence was significantly higher in ICI-exposed subjects (OR, 1.64; 95% CI, 1.01-2.67). Risks of oxygen dependence (OR, 1.13; 95% CI, 0.75-1.73), kidney injury (OR, 1.15; 95% CI, 0.77-1.71), and myocardial injury (OR, 1.76; 95% CI, 1.00-3.10) were not significantly different. There was no difference between groups in the time to hospital discharge alive (p = 0.62). CONCLUSIONS Exposure to ICIs within 6 months before high-risk surgery was not associated with the composite outcome of cardiopulmonary instability or organ injury in patients with cancer. The potential for an association with the secondary outcomes of cardiac instability and injury is worthy of future study.
Collapse
Affiliation(s)
- Ying-Hung Tang
- Department of Anesthesiology, Mackay Memorial Hospital, Taipei, Taiwan
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
| | - Jules Bergmann
- Department of Anesthesiology/Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Dhananjay Vaidya
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Nauder Faraday
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
- Department of Anesthesiology/Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
7
|
Rodriguez-Berriguete G, Puliyadi R, Machado N, Barberis A, Prevo R, McLaughlin M, Buffa FM, Harrington KJ, Higgins GS. Antitumour effect of the mitochondrial complex III inhibitor Atovaquone in combination with anti-PD-L1 therapy in mouse cancer models. Cell Death Dis 2024; 15:32. [PMID: 38212297 PMCID: PMC10784292 DOI: 10.1038/s41419-023-06405-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 12/14/2023] [Accepted: 12/20/2023] [Indexed: 01/13/2024]
Abstract
Immune checkpoint blockade (ICB) provides effective and durable responses for several tumour types by unleashing an immune response directed against cancer cells. However, a substantial number of patients treated with ICB develop relapse or do not respond, which has been partly attributed to the immune-suppressive effect of tumour hypoxia. We have previously demonstrated that the mitochondrial complex III inhibitor atovaquone alleviates tumour hypoxia both in human xenografts and in cancer patients by decreasing oxygen consumption and consequently increasing oxygen availability in the tumour. Here, we show that atovaquone alleviates hypoxia and synergises with the ICB antibody anti-PD-L1, significantly improving the rates of tumour eradication in the syngeneic CT26 model of colorectal cancer. The synergistic effect between atovaquone and anti-PD-L1 relied on CD8+ T cells, resulted in the establishment of a tumour-specific memory immune response, and was not associated with any toxicity. We also tested atovaquone in combination with anti-PD-L1 in the LLC (lung) and MC38 (colorectal) cancer syngeneic models but, despite causing a considerable reduction in tumour hypoxia, atovaquone did not add any therapeutic benefit to ICB in these models. These results suggest that atovaquone has the potential to improve the outcomes of patients treated with ICB, but predictive biomarkers are required to identify individuals likely to benefit from this intervention.
Collapse
Affiliation(s)
| | - Rathi Puliyadi
- Department of Oncology, University of Oxford, Oxford, UK
| | - Nicole Machado
- Department of Oncology, University of Oxford, Oxford, UK
| | | | - Remko Prevo
- Department of Oncology, University of Oxford, Oxford, UK
| | | | - Francesca M Buffa
- Department of Oncology, University of Oxford, Oxford, UK
- Department of Computing Sciences, Bocconi University, Milan, Italy
| | | | | |
Collapse
|
8
|
Ebinama U, Sheshadri A, Anand K, Swaminathan I. Pulmonary Immune-Related Adverse Events of PD-1 Versus PD-L1 Checkpoint Inhibitors: A Retrospective Review of Pharmacovigilance. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2023; 6:177-184. [PMID: 38143955 PMCID: PMC10734392 DOI: 10.36401/jipo-22-38] [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: 12/31/2022] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 12/26/2023]
Abstract
Introduction Immune checkpoint inhibitors (ICIs) have revolutionized cancer therapeutics. However, immune-related adverse events (irAEs) increase morbidity and mortality and thereby limit therapeutic utility. The real-world incidence of the entire spectrum of pulmonary irAEs has not been systematically described. The objective of this study is to assess the risk of developing pulmonary irAEs (pneumonitis, pleural events [i.e., effusion and pleurisy], exacerbations of airway disease [i.e., bronchitis and bronchiectasis], and sarcoidosis) with exposure to five commonly used ICIs: nivolumab, pembrolizumab, durvalumab, avelumab, and atezolizumab. Methods We conducted a retrospective review of the Food and Drug Administration Adverse Events Reporting System (FAERS) pharmacovigilance database. We collected data from 2012 to 2021 to assess the risk of pulmonary irAEs and performed a disproportionality analysis using Open-Vigil, a software package used for analysis of pharmacovigilance data, to calculate reporting odds ratios (RORs). We used 95% CIs to evaluate the precision of RORs. An ROR greater than 1 and the upper limit of the 95% CI indicated statistical significance. Results A total of 17,273,403 events were reported in FAERS between 2012 and 2021. Of these, 88,099 (0.5%) were attributed to the PD-1 (programmed cell death protein 1) inhibitors and 21,905 (0.1%) to PD-L1 (programmed death ligand 1) inhibitors of interest. The most common indication for using the ICIs of interest was lung cancer: a total of 2832 (46.70%) for the PD-1 inhibitors and 1311 (70.9%) for the PD-L1 inhibitors. In the anti-PD-1 group, 2342 (38.6%) patients were hospitalized, and 1962 (32.4%) patients died from the lung adverse event. In the PD-L1 group, 744 (40.3%) patients were hospitalized, and 520 (28.1%) patients died from the event. Nivolumab resulted in the highest statistically significant risk (ROR, 10.5; 95% CI, 10.1-10.9) for pneumonitis. Avelumab had a lesser risk for pneumonitis (ROR, 0.2; 95% CI, 0.2-0.3). The risk for pleural events was highest with nivolumab (ROR, 3.6; 95% CI, 3.4-3.9), followed by pembrolizumab (ROR, 1.8; 95% CI; 1.6-2.0) (p < 0.001), with the lowest risks from durvalumab, atezolizumab, and avelumab. For ICI-related sarcoidosis, the risk was most significant with pembrolizumab (ROR, 3.6; 95% CI, 2.8-4.7), followed by nivolumab (ROR, 2.5; 95% CI, 1.9-3.5) (p < 0.001). The RORs for all five ICIs were less than 1 for exacerbations of airway diseases as compared with other drugs. Conclusion Using a pharmacovigilance database, we found an increased risk of multiple pulmonary irAEs after ICI therapy, particularly with PD-1 inhibitors. Further work is needed to investigate the incidence of pulmonary irAEs other than pneumonitis.
Collapse
Affiliation(s)
- Ugochi Ebinama
- Department of Internal Medicine, The University of Texas Health Sciences Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Ajay Sheshadri
- Department of Pulmonary Medicine, Division of Internal Medicine, Houston, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kartik Anand
- Callahan Cancer Center, Great Plains Health, North Platte, NE, USA
- Mission Cancer and Blood, Des Moines, IA, USA
| | - Iyer Swaminathan
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
9
|
Sacchi de Camargo Correia G, Pai T, Li S, Connor D, Zhao Y, Lou Y, Manochakian R. Immune-Related Adverse Events in Patients with Lung Cancer. Curr Oncol Rep 2023; 25:1259-1275. [PMID: 37782426 DOI: 10.1007/s11912-023-01462-w] [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] [Accepted: 08/30/2023] [Indexed: 10/03/2023]
Abstract
PURPOSE OF REVIEW Immune checkpoint inhibitors (ICI) have been widely adopted for the treatment of lung cancer since receiving first U.S Food and Drug Administration (FDA) approval in 2015. However, along with their use, the occurrence of immune-related adverse events (irAEs) has presented a challenge for both patients and oncology providers. In this manuscript, we reviewed the clinical trials that led to the approval of ICI by the FDA for the treatment of lung cancer between 2015 and 2023 to establish the frequency of irAEs in this patient population. Among the adverse events associated with ICI, we focused on the most common and relevant ones, including hypothyroidism, pneumonitis, diarrhea/colitis, skin rash, and hepatitis. RECENT FINDINGS We thoroughly examined the available literature, including society guidelines concerning these complications, to discuss various aspects such as their pathophysiology, epidemiology, diagnostic process, grading system, and clinical management. Additionally, we explored the association between irAEs and disease response. The management of irAEs is a crucial aspect of oncologic care, particularly due to their potential to cause severe and life-threatening clinical manifestations. We present each pertinent aspect in a concise and organized manner to provide guidance and assistance to oncology providers managing these patients in both outpatient and inpatient settings.
Collapse
Affiliation(s)
| | - Tanmayi Pai
- Division of Hematology and Medical Oncology, Mayo Clinic, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA
| | - Shenduo Li
- Division of Hematology and Medical Oncology, Mayo Clinic, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA
| | - Dana Connor
- Division of Hematology and Medical Oncology, Mayo Clinic, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA
| | - Yujie Zhao
- Division of Hematology and Medical Oncology, Mayo Clinic, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA
| | - Yanyan Lou
- Division of Hematology and Medical Oncology, Mayo Clinic, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA
| | - Rami Manochakian
- Division of Hematology and Medical Oncology, Mayo Clinic, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA.
| |
Collapse
|
10
|
Vithayathil M, D'Alessio A, Fulgenzi CAM, Nishida N, Schönlein M, von Felden J, Schulze K, Wege H, Saeed A, Wietharn B, Hildebrand H, Wu L, Ang C, Marron TU, Weinmann A, Galle PR, Bettinger D, Bengsch B, Vogel A, Balcar L, Scheiner B, Lee PC, Huang YH, Amara S, Muzaffar M, Naqash AR, Cammarota A, Zanuso V, Pressiani T, Pinter M, Cortellini A, Kudo M, Rimassa L, Pinato DJ, Sharma R. Impact of body mass index in patients receiving atezolizumab plus bevacizumab for hepatocellular carcinoma. Hepatol Int 2023; 17:904-914. [PMID: 37005953 PMCID: PMC10386929 DOI: 10.1007/s12072-023-10491-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 01/16/2023] [Indexed: 04/04/2023]
Abstract
BACKGROUND Atezolizumab plus bevacizumab (Atezo/Bev) is first line-treatment for unresectable hepatocellular carcinoma (HCC). Body mass index (BMI) has demonstrated predictive value for response to immunotherapy in non-HCC cancer types. Our study investigated the effect of BMI on safety and efficacy of real-life use of Atezo/Bev for unresectable HCC. METHODS 191 consecutive patients from seven centres receiving Atezo/Bev were included in the retrospective study. Overall survival (OS), progression-free survival (PFS), overall response rate (ORR) and disease control rate (DCR) defined by RECIST v1.1 were measured in overweight (BMI ≥ 25) and non-overweight (BMI < 25) patients. Treatment-related adverse events (trAEs) were evaluated. RESULTS Patients in the overweight cohort (n = 94) had higher rates of non-alcoholic fatty liver disease (NAFLD) and lower rates of Hepatitis B compared to non-overweight cohort (n = 97). Baseline Child-Pugh class and Barcelona Clinic Liver Cancer stage were similar between cohorts, with lower rates of extrahepatic spread in the overweight group. Overweight patients had similar OS compared to non-overweight (median OS 15.1 vs. 14.9 months; p = 0.99). BMI did not influence median PFS (7.1 vs. 6.1 months; p = 0.42), ORR (27.2% vs. 22.0%; p = 0.44) and DCR (74.1% vs. 71.9%; p = 0.46). There were higher rates of atezolizumab-related fatigue (22.3% vs. 10.3%; p = 0.02) and bevacizumab-related thrombosis (8.5% vs. 2.1%; p = 0.045) in the overweight patients, but overall trAEs and treatment discontinuation were comparable between cohorts. CONCLUSION Atezo/Bev has comparable efficacy in overweight HCC patients, with an increase in treatment-related fatigue and thrombosis. Combination therapy is safe and efficacious to use in overweight patients, including those with underlying NAFLD.
Collapse
Affiliation(s)
- Mathew Vithayathil
- Department of Surgery & Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK
| | - Antonio D'Alessio
- Department of Surgery & Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Claudia Angela Maria Fulgenzi
- Department of Surgery & Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK
- Division of Medical Oncology, Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Naoshi Nishida
- Department of Gastroenterology and Hepatology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Martin Schönlein
- Department of Oncology, Hematology and Bone Marrow Transplantation With Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Johann von Felden
- Department of Gastroenterology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kornelius Schulze
- Department of Gastroenterology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Henning Wege
- Department of Gastroenterology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anwaar Saeed
- Division of Medical Oncology, Department of Medicine, Kansas University Cancer Center, Kansas City, KS, USA
| | - Brooke Wietharn
- Division of Medical Oncology, Department of Medicine, Kansas University Cancer Center, Kansas City, KS, USA
| | - Hannah Hildebrand
- Division of Medical Oncology, Department of Medicine, Kansas University Cancer Center, Kansas City, KS, USA
| | - Linda Wu
- Division of Hematology/Oncology, Department of Medicine, Tisch Cancer Institute, Mount Sinai Hospital, New York, NY, USA
| | - Celina Ang
- Division of Hematology/Oncology, Department of Medicine, Tisch Cancer Institute, Mount Sinai Hospital, New York, NY, USA
| | - Thomas U Marron
- Division of Hematology/Oncology, Department of Medicine, Tisch Cancer Institute, Mount Sinai Hospital, New York, NY, USA
| | - Arndt Weinmann
- I. Medical Department, University Medical Center Mainz, Mainz, Germany
| | - Peter R Galle
- I. Medical Department, University Medical Center Mainz, Mainz, Germany
| | - Dominik Bettinger
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Faculty of Medicine, Freiburg University Medical Center, University of Freiburg, Freiburg, Germany
| | - Bertram Bengsch
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Faculty of Medicine, Freiburg University Medical Center, University of Freiburg, Freiburg, Germany
- University of Freiburg, Signalling Research Centers BIOSS and CIBSS, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site, Freiburg, Germany
| | | | - Lorenz Balcar
- Division of Gastroenterology & Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Bernhard Scheiner
- Division of Gastroenterology & Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Pei-Chang Lee
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Hsiang Huang
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Suneetha Amara
- Division of Hematology/Oncology, East Carolina University, Greenville, NC, USA
| | - Mahvish Muzaffar
- Division of Hematology/Oncology, East Carolina University, Greenville, NC, USA
| | - Abdul Rafeh Naqash
- Division of Hematology/Oncology, East Carolina University, Greenville, NC, USA
- Medical Oncology/TSET Phase 1 Program, Stephenson Cancer Center, University of Oklahoma, Norman, OK, USA
| | - Antonella Cammarota
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Valentina Zanuso
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Tiziana Pressiani
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Matthias Pinter
- Division of Gastroenterology & Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Alessio Cortellini
- Department of Surgery & Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Lorenza Rimassa
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - David J Pinato
- Department of Surgery & Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK
- Division of Oncology, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Rohini Sharma
- Department of Surgery & Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK.
| |
Collapse
|
11
|
Celikdemir B, Houben R, Kervarrec T, Samimi M, Schrama D. Current and preclinical treatment options for Merkel cell carcinoma. Expert Opin Biol Ther 2023; 23:1015-1034. [PMID: 37691397 DOI: 10.1080/14712598.2023.2257603] [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: 03/28/2023] [Revised: 09/04/2023] [Accepted: 09/07/2023] [Indexed: 09/12/2023]
Abstract
INTRODUCTION Merkel cell carcinoma (MCC) is a rare, highly aggressive form of skin cancer with neuroendocrine features. The origin of this cancer is still unclear, but research in the last 15 years has demonstrated that MCC arises via two distinct etiologic pathways, i.e. virus and UV-induced. Considering the high mortality rate and the limited therapeutic options available, this review aims to highlight the significance of MCC research and the need for advancement in MCC treatment. AREAS COVERED With the advent of the immune checkpoint inhibitor therapies, we now have treatment options providing a survival benefit for patients with advanced MCC. However, the issue of primary and acquired resistance to these therapies remains a significant concern. Therefore, ongoing efforts seeking additional therapeutic targets and approaches for MCC therapy are a necessity. Through a comprehensive literature search, we provide an overview on recent preclinical and clinical studies with respect to MCC therapy. EXPERT OPINION Currently, the only evidence-based therapy for MCC is immune checkpoint blockade with anti-PD-1/PD-L1 for advanced patients. Neoadjuvant, adjuvant and combined immune checkpoint blockade are promising treatment options.
Collapse
Affiliation(s)
- Büke Celikdemir
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - Roland Houben
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - Thibault Kervarrec
- Department of Pathology, Centre Hospitalier Universitaire De Tours, Tours, France
| | - Mahtab Samimi
- Department of Dermatology, University Hospital of Tours, Tours, France
| | - David Schrama
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
12
|
Kleczko EK, Nguyen DT, Marsh KH, Bauer CD, Li AS, Monaghan MLT, Berger MD, Furgeson SB, Gitomer BY, Chonchol MB, Clambey ET, Zimmerman KA, Nemenoff RA, Hopp K. Immune checkpoint activity regulates polycystic kidney disease progression. JCI Insight 2023; 8:e161318. [PMID: 37345660 PMCID: PMC10371237 DOI: 10.1172/jci.insight.161318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/12/2023] [Indexed: 06/23/2023] Open
Abstract
Innate and adaptive immune cells modulate the severity of autosomal dominant polycystic kidney disease (ADPKD), a common kidney disease with inadequate treatment options. ADPKD has parallels with cancer, in which immune checkpoint inhibitors have been shown to reactivate CD8+ T cells and slow tumor growth. We have previously shown that in PKD, CD8+ T cell loss worsens disease. This study used orthologous early-onset and adult-onset ADPKD models (Pkd1 p.R3277C) to evaluate the role of immune checkpoints in PKD. Flow cytometry of kidney cells showed increased levels of programmed cell death protein 1 (PD-1)/cytotoxic T lymphocyte associated protein 4 (CTLA-4) on T cells and programmed cell death ligand 1 (PD-L1)/CD80 on macrophages and epithelial cells in Pkd1RC/RC mice versus WT, paralleling disease severity. PD-L1/CD80 was also upregulated in ADPKD human cells and patient kidney tissue versus controls. Genetic PD-L1 loss or treatment with an anti-PD-1 antibody did not impact PKD severity in early-onset or adult-onset ADPKD models. However, treatment with anti-PD-1 plus anti-CTLA-4, blocking 2 immune checkpoints, improved PKD outcomes in adult-onset ADPKD mice; neither monotherapy altered PKD severity. Combination therapy resulted in increased kidney CD8+ T cell numbers/activation and decreased kidney regulatory T cell numbers correlative with PKD severity. Together, our data suggest that immune checkpoint activation is an important feature of and potential novel therapeutic target in ADPKD.
Collapse
Affiliation(s)
- Emily K. Kleczko
- Department of Medicine, Division of Renal Diseases and Hypertension
| | - Dustin T. Nguyen
- Department of Medicine, Division of Renal Diseases and Hypertension
| | - Kenneth H. Marsh
- Department of Medicine, Division of Renal Diseases and Hypertension
| | - Colin D. Bauer
- Department of Medicine, Division of Renal Diseases and Hypertension
| | - Amy S. Li
- Department of Medicine, Division of Renal Diseases and Hypertension
| | | | | | - Seth B. Furgeson
- Department of Medicine, Division of Renal Diseases and Hypertension
| | | | - Michel B. Chonchol
- Department of Medicine, Division of Renal Diseases and Hypertension
- Consortium for Fibrosis Research and Translation, and
| | - Eric T. Clambey
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kurt A. Zimmerman
- Department of Internal Medicine, Division of Nephrology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Raphael A. Nemenoff
- Department of Medicine, Division of Renal Diseases and Hypertension
- Consortium for Fibrosis Research and Translation, and
| | - Katharina Hopp
- Department of Medicine, Division of Renal Diseases and Hypertension
- Consortium for Fibrosis Research and Translation, and
| |
Collapse
|
13
|
Kanbayashi Y, Shimizu T, Anzai M, Kawai R, Uchida M. Evaluation of Cardiac Adverse Events with Nivolumab Using a Japanese Real-World Database. Clin Drug Investig 2023; 43:177-184. [PMID: 36780109 DOI: 10.1007/s40261-023-01246-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2023] [Indexed: 02/14/2023]
Abstract
BACKGROUND Nivolumab has been used for the treatment of various types of cancers and has achieved improvements in overall survival. However, nivolumab can cause a variety of adverse events (AEs). Among these, cardiac-specific AEs have received little attention in clinical trials, despite their life-threatening potential. OBJECTIVE The present study aimed to determine the risk of nivolumab-induced cardiac AEs, time to onset, incidence rates, and post hoc outcomes using the Japanese Adverse Drug Event Report database. METHODS We analyzed data for the period between April 2004 and March 2021. Data on cardiac AEs were extracted and relative risk of AEs was estimated using the reporting odds ratio (ROR). RESULTS We analyzed 1,772,494 reports and identified 18,721 reports of AEs caused by nivolumab. Of these, 409 reports involved cardiac AEs. Signals were detected for four cardiac AEs: myocarditis; pericardial effusion; pericarditis; and immune-mediated myocarditis. Among these, myocarditis was the most frequently reported (35.0%) and included fatal cases. A histogram of times to onset showed nivolumab-associated AEs occurring 41-127 days after starting administration, with outlier cases of myocarditis or pericardial effusion occurring after more than one year, both with catastrophic consequences. CONCLUSION This study focused on cardiac AEs caused by nivolumab as post-marketing AEs. Myocarditis and pericardial effusion have been associated with some fatal cases after administration of nivolumab. Patients should be monitored for signs of onset for these AEs, not only at the start of administration, but also over an extended period after nivolumab administration.
Collapse
Affiliation(s)
- Yuko Kanbayashi
- Department of Education and Research Center for Clinical Pharmacy, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan.
| | - Tadashi Shimizu
- School of Pharmacy, Hyogo Medical University, 1-3-6 Minatojima, Kobe, Hyogo, 650-8530, Japan
| | - Miku Anzai
- Department of Education and Research Center for Pharmacy Practice, Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, 97-1 Kodominamihokotate, Kyotanabe, Kyoto, 610-0395, Japan
| | - Rika Kawai
- Department of Education and Research Center for Pharmacy Practice, Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, 97-1 Kodominamihokotate, Kyotanabe, Kyoto, 610-0395, Japan
| | - Mayako Uchida
- Department of Education and Research Center for Pharmacy Practice, Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, 97-1 Kodominamihokotate, Kyotanabe, Kyoto, 610-0395, Japan
| |
Collapse
|
14
|
Heeg B, Verhoek A, Tremblay G, Harari O, Soltanifar M, Chu H, Roychoudhury S, Cappelleri JC. Bayesian hierarchical model-based network meta-analysis to overcome survival extrapolation challenges caused by data immaturity. J Comp Eff Res 2023; 12:e220159. [PMID: 36651607 PMCID: PMC10288968 DOI: 10.2217/cer-2022-0159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 12/21/2022] [Indexed: 01/19/2023] Open
Abstract
Aim: This research evaluated standard Weibull mixture cure (WMC) network meta-analysis (NMA) with Bayesian hierarchical (BH) WMC NMA to inform long-term survival of therapies. Materials & methods: Four trials in previously treated metastatic non-small-cell lung cancer with PD-L1 >1% were used comparing docetaxel with nivolumab, pembrolizumab and atezolizumab. Cure parameters related to a certain treatment class were assumed to share a common distribution. Results: Standard WMC NMA predicted cure rates were 0.03 (0.01; 0.07), 0.18 (0.12; 0.24), 0.07 (0.02; 0.15) and 0.03 (0.00; 0.09) for docetaxel, nivolumab, pembrolizumab and atezolizumab, respectively, with corresponding incremental life years (LY) of 3.11 (1.65; 4.66), 1.06 (0.41; 2.37) and 0.42 (-0.57; 1.68). The Bayesian hierarchical-WMC-NMA rates were 0.06 (0.03; 0.10), 0.17 (0.11; 0.23), 0.12 (0.05; 0.20) and 0.12 (0.03; 0.23), respectively, with incremental LY of 2.35 (1.04; 3.93), 1.67 (0.68; 2.96) and 1.36 (-0.05; 3.64). Conclusion: BH-WMC-NMA impacts incremental mean LYs and cost-effectiveness ratios, potentially affecting reimbursement decisions.
Collapse
Affiliation(s)
- Bart Heeg
- Cytel RWAA, Weena 316, 3012 NJ, Rotterdam, The Netherlands
| | - Andre Verhoek
- Cytel RWAA, Weena 316, 3012 NJ, Rotterdam, The Netherlands
| | | | | | | | - Haitao Chu
- Pfizer Inc, 445 Eastern Point Road, MS 8260-2502, Groton, CT 06340, USA
| | - Satrajit Roychoudhury
- Pfizer Inc, 445 Eastern Point Road, MS 8260-2502, Groton, CT 06340, USA
- Pfizer Inc., 235 E 42nd St, New York, NY 10017, USA
| | | |
Collapse
|
15
|
Li M, Quintana A, Alberts E, Hung MS, Boulat V, Ripoll MM, Grigoriadis A. B Cells in Breast Cancer Pathology. Cancers (Basel) 2023; 15:1517. [PMID: 36900307 PMCID: PMC10000926 DOI: 10.3390/cancers15051517] [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: 12/22/2022] [Revised: 02/13/2023] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
B cells have recently become a focus in breast cancer pathology due to their influence on tumour regression, prognosis, and response to treatment, besides their contribution to antigen presentation, immunoglobulin production, and regulation of adaptive responses. As our understanding of diverse B cell subsets in eliciting both pro- and anti-inflammatory responses in breast cancer patients increases, it has become pertinent to address the molecular and clinical relevance of these immune cell populations within the tumour microenvironment (TME). At the primary tumour site, B cells are either found spatially dispersed or aggregated in so-called tertiary lymphoid structures (TLS). In axillary lymph nodes (LNs), B cell populations, amongst a plethora of activities, undergo germinal centre reactions to ensure humoral immunity. With the recent approval for the addition of immunotherapeutic drugs as a treatment option in the early and metastatic settings for triple-negative breast cancer (TNBC) patients, B cell populations or TLS may resemble valuable biomarkers for immunotherapy responses in certain breast cancer subgroups. New technologies such as spatially defined sequencing techniques, multiplex imaging, and digital technologies have further deciphered the diversity of B cells and the morphological structures in which they appear in the tumour and LNs. Thus, in this review, we comprehensively summarise the current knowledge of B cells in breast cancer. In addition, we provide a user-friendly single-cell RNA-sequencing platform, called "B singLe cEll rna-Seq browSer" (BLESS) platform, with a focus on the B cells in breast cancer patients to interrogate the latest publicly available single-cell RNA-sequencing data collected from diverse breast cancer studies. Finally, we explore their clinical relevance as biomarkers or molecular targets for future interventions.
Collapse
Affiliation(s)
- Mengyuan Li
- Cancer Bioinformatics, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 9RT, UK
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 9RT, UK
| | | | - Elena Alberts
- Cancer Bioinformatics, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 9RT, UK
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 9RT, UK
- Immunity and Cancer Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Miu Shing Hung
- Cancer Bioinformatics, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 9RT, UK
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 9RT, UK
| | - Victoire Boulat
- Cancer Bioinformatics, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 9RT, UK
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 9RT, UK
- Immunity and Cancer Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Mercè Martí Ripoll
- Immunology Unit, Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
- Biosensing and Bioanalysis Group, Institute of Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Anita Grigoriadis
- Cancer Bioinformatics, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 9RT, UK
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 9RT, UK
- Breast Cancer Now Unit, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 9RT, UK
| |
Collapse
|
16
|
Barsheshet Y, Voloshin T, Brant B, Cohen G, Koren L, Blatt R, Cahal S, Haj Khalil T, Zemer Tov E, Paz R, Klein-Goldberg A, Tempel-Brami C, Jacobovitch S, Volodin A, Kan T, Koltun B, David C, Haber A, Giladi M, Weinberg U, Palti Y. Tumor Treating Fields (TTFields) Concomitant with Immune Checkpoint Inhibitors Are Therapeutically Effective in Non-Small Cell Lung Cancer (NSCLC) In Vivo Model. Int J Mol Sci 2022; 23:ijms232214073. [PMID: 36430552 PMCID: PMC9696536 DOI: 10.3390/ijms232214073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/06/2022] [Accepted: 11/10/2022] [Indexed: 11/17/2022] Open
Abstract
Tumor Treating Fields (TTFields) are electric fields that exert physical forces to disrupt cellular processes critical for cancer cell viability and tumor progression. TTFields induce anti-mitotic effects through the disruption of the mitotic spindle and abnormal chromosome segregation, which trigger several forms of cell death, including immunogenic cell death (ICD). The efficacy of TTFields concomitant with anti-programmed death-1 (anti-PD-1) treatment was previously shown in vivo and is currently under clinical investigation. Here, the potential of TTFields concomitant with anti- PD-1/anti-cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA-4) or anti-programmed death-ligand 1 (anti-PD-L1) immune checkpoint inhibitors (ICI) to improve therapeutic efficacy was examined in lung tumor-bearing mice. Increased circulating levels of high mobility group box 1 protein (HMGB1) and elevated intratumoral levels of phosphorylated eukaryotic translation initiation factor 2α (p-eIF2α) were found in the TTFields-treated mice, indicative of ICD induction. The concomitant application of TTFields and ICI led to a significant decrease in tumor volume as compared to all other groups. In addition, significant increases in the number of tumor-infiltrating immune cells, specifically cytotoxic T-cells, were observed in the TTFields plus anti-PD-1/anti-CTLA-4 or anti-PD-L1 groups. Correspondingly, cytotoxic T-cells isolated from these tumors showed higher levels of IFN-γ production. Collectively, these results suggest that TTFields have an immunoactivating role that may be leveraged for concomitant treatment with ICI to achieve better tumor control by enhancing antitumor immunity.
Collapse
|
17
|
Daly RJ, Scott AM, Klein O, Ernst M. Enhancing therapeutic anti-cancer responses by combining immune checkpoint and tyrosine kinase inhibition. Mol Cancer 2022; 21:189. [PMID: 36175961 PMCID: PMC9523960 DOI: 10.1186/s12943-022-01656-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 09/19/2022] [Indexed: 11/10/2022] Open
Abstract
Over the past decade, immune checkpoint inhibitor (ICI) therapy has been established as the standard of care for many types of cancer, but the strategies employed have continued to evolve. Recently, much clinical focus has been on combining targeted therapies with ICI for the purpose of manipulating the immune setpoint. The latter concept describes the equilibrium between factors that promote and those that suppress anti-cancer immunity. Besides tumor mutational load and other cancer cell-intrinsic determinants, the immune setpoint is also governed by the cells of the tumor microenvironment and how they are coerced by cancer cells to support the survival and growth of the tumor. These regulatory mechanisms provide therapeutic opportunities to intervene and reduce immune suppression via application of small molecule inhibitors and antibody-based therapies against (receptor) tyrosine kinases and thereby improve the response to ICIs. This article reviews how tyrosine kinase signaling in the tumor microenvironment can promote immune suppression and highlights how therapeutic strategies directed against specific tyrosine kinases can be used to lower the immune setpoint and elicit more effective anti-tumor immunity.
Collapse
Affiliation(s)
- Roger J Daly
- Cancer Program, Monash Biomedicine Discovery Institute, Monash University, 23 Innovation Walk, Clayton, VIC, 3800, Australia.
- Department of Biochemistry & Molecular Biology, Monash University, 23 Innovation Walk, Clayton, VIC, 3800, Australia.
| | - Andrew M Scott
- Department of Biochemistry & Molecular Biology, Monash University, 23 Innovation Walk, Clayton, VIC, 3800, Australia
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, 145 Studley Rd, Melbourne-Heidelberg, VIC, 3084, Australia
- Department of Molecular Imaging & Therapy, Austin Health, and Faculty of Medicine, University of Melbourne, 145 Studley Rd, Melbourne-Heidelberg, VIC, 3084, Australia
| | - Oliver Klein
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, 145 Studley Rd, Melbourne-Heidelberg, VIC, 3084, Australia
| | - Matthias Ernst
- Department of Biochemistry & Molecular Biology, Monash University, 23 Innovation Walk, Clayton, VIC, 3800, Australia.
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, 145 Studley Rd, Melbourne-Heidelberg, VIC, 3084, Australia.
| |
Collapse
|
18
|
Chan P, Peskov K, Song X. Applications of Model-Based Meta-Analysis in Drug Development. Pharm Res 2022; 39:1761-1777. [PMID: 35174432 PMCID: PMC9314311 DOI: 10.1007/s11095-022-03201-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/11/2022] [Indexed: 12/13/2022]
Abstract
Model-based meta-analysis (MBMA) is a quantitative approach that leverages published summary data along with internal data and can be applied to inform key drug development decisions, including the benefit-risk assessment of a treatment under investigation. These risk-benefit assessments may involve determining an optimal dose compared against historic external comparators of a particular disease indication. MBMA can provide a flexible framework for interpreting aggregated data from historic reference studies and therefore should be a standard tool for the model-informed drug development (MIDD) framework.In addition to pairwise and network meta-analyses, MBMA provides further contributions in the quantitative approaches with its ability to incorporate longitudinal data and the pharmacologic concept of dose-response relationship, as well as to combine individual- and summary-level data and routinely incorporate covariates in the analysis.A common application of MBMA is the selection of optimal dose and dosing regimen of the internal investigational molecule to evaluate external benchmarking and to support comparator selection. Two case studies provided examples in applications of MBMA in biologics (durvalumab + tremelimumab for safety) and small molecule (fenebrutinib for efficacy) to support drug development decision-making in two different but well-studied disease areas, i.e., oncology and rheumatoid arthritis, respectively.Important to the future directions of MBMA include additional recognition and engagement from drug development stakeholders for the MBMA approach, stronger collaboration between pharmacometrics and statistics, expanded data access, and the use of machine learning for database building. Timely, cost-effective, and successful application of MBMA should be part of providing an integrated view of MIDD.
Collapse
Affiliation(s)
- Phyllis Chan
- Clinical Pharmacology, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA.
| | - Kirill Peskov
- M&S Decisions LLC, Moscow, Russia
- Sechenov First Moscow State Medical University, Moscow, Russia
- STU 'Sirius', Sochi, Russia
| | - Xuyang Song
- Clinical Pharmacology and Quantitative Pharmacology, AstraZeneca, 1 Medimmune Way, Gaithersburg, MD, 20878, USA
| |
Collapse
|
19
|
Banna GL, Cantale O, Muthuramalingam S, Cave J, Comins C, Cortellini A, Addeo A, Signori A, McKenzie H, Escriu C, Barone G, Chan S, Hicks A, Bainbridge H, Pinato DJ, Ottensmeier C, Gomes F. Efficacy outcomes and prognostic factors from real-world patients with advanced non-small-cell lung cancer treated with first-line chemoimmunotherapy: The Spinnaker retrospective study. Int Immunopharmacol 2022; 110:108985. [PMID: 35777264 DOI: 10.1016/j.intimp.2022.108985] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/14/2022] [Accepted: 06/16/2022] [Indexed: 12/18/2022]
Abstract
BACKGROUND Efficacy outcomes and prognostic factors of real-world patients with advanced non-small cell lung cancer (aNSCLC) treated with first-line chemoimmunotherapy are still limited. PATIENTS AND METHODS In the retrospective Spinnaker study, data was collected from patients in six United Kingdom and one Swiss oncology centres with first-line pembrolizumab plus platinum-based chemotherapy. Efficacy outcomes and potential prognostic factors were estimated aiming at developing a prognostic model. RESULTS Three-hundred-eight patients were included, 32% ≥ 70 years, with ≥ 3 metastatic sites in 33%, brain or liver metastases in 10% and 12%, respectively. With a median follow-up of 18.0 months (mo.) (range, 15.9-20.1), median overall survival (OS) and progression-free survival (PFS) were 12.7 mo. (range, 10.2-15.2), and 8.0 mo. (range, 7.1-8.8), respectively. The neutrophils-to-lymphocytes ratio (NLR) and systemic immune-inflammatory index (SII) (i.e., NLR × platelet count) were both significantly higher in ECOG PS 1 (p = 0.0147 and p = 0.0018, respectively), underweight or normal body mass index (p = 0.0456 and p = 0.0062, respectively), ≥3 metastatic sites (p = 0.0069 and p = 0.112), pretreatment steroids (p = 0.0019 and p = 0.0017). By MVA, the number of metastatic sites ≥ 3 (p < 0.001 and p = 0.002), squamous histology (p = 0.033 and p = 0.013) and SII ≥ 1444 (p = 0.031 and p = 0.009, respectively) were associated with both worse OS and PFS and led to a highly discriminating three-class risk prognostic model. CONCLUSION Real-world PFS with chemoimmunotherapy in aNSCLC patients is similar to that reported in clinical trials. A high number of metastatic sites, squamous histology and high SII are adverse prognostic factors that might contribute to a clinically useful prognostic model.
Collapse
Affiliation(s)
| | - Ornella Cantale
- Medical Oncology Department, University of Turin, San Luigi Gonzaga Hospital, Orbassano, Italy
| | | | | | | | | | | | | | | | - Carles Escriu
- The Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool, UK
| | - Gloria Barone
- United Lincolnshire Hospitals NHS Trust, Lincoln, UK
| | - Samuel Chan
- Portsmouth Hospitals University NHS Trust, Portsmouth, UK
| | | | | | - David J Pinato
- Imperial College London, Hammersmith Hospital, London, UK; Division of Oncology, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Christian Ottensmeier
- Clatterbridge Cancer Centre NHS Foundation Trust, University of Liverpool, Liverpool, UK
| | - Fabio Gomes
- The Christie NHS Foundation Trust, Manchester, UK
| |
Collapse
|
20
|
Chen Y, Lai X. Modeling the effect of gut microbiome on therapeutic efficacy of immune checkpoint inhibitors against cancer. Math Biosci 2022; 350:108868. [PMID: 35753521 DOI: 10.1016/j.mbs.2022.108868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 11/25/2022]
Abstract
Immune checkpoint inhibitors have been shown to be highly successful against some solid metastatic malignancies, but only for a subset of patients who show durable clinical responses. The overall patient response rate is limited due to the interpatient heterogeneity. Preclinical and clinical studies have recently shown that the therapeutic responses can be improved through the modulation of gut microbiome. However, the underlying mechanisms are not fully understood. In this paper, we explored the effect of favorable and unfavorable gut bacteria on the therapeutic efficacy of anti-PD-1 against cancer by modeling the tumor-immune-gut microbiome interactions, and further examined the predictive markers of responders and non-responders to anti-PD-1. The dynamics of the gut bacteria was fitted to the clinical data of melanoma patients, and virtual patients data were generated based on the clinical patient survival data. Our simulation results show that low initial growth rate and low level of favorable bacteria at the initiation of anti-PD-1 therapy are predictive of non-responders, while high level of favorable bacteria at the initiation of anti-PD-1 therapy is predictive of responders. Simulation results also confirmed that it is possible to promote patients' response rate to anti-PD-1 by manipulating the gut bacteria composition of non-responders, whereby achieving long-term progression-free survival.
Collapse
Affiliation(s)
- Yu Chen
- Institute for Mathematical Sciences, Renmin University of China, Beijing, 100872, China
| | - Xiulan Lai
- Institute for Mathematical Sciences, Renmin University of China, Beijing, 100872, China.
| |
Collapse
|
21
|
Suraya R, Tachihara M, Nagano T, Nishimura Y, Kobayashi K. Immunotherapy in Advanced Non-Small Cell Lung Cancers: Current Status and Updates. Cancer Manag Res 2022; 14:2079-2090. [PMID: 35769229 PMCID: PMC9234310 DOI: 10.2147/cmar.s366738] [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: 03/16/2022] [Accepted: 06/02/2022] [Indexed: 12/04/2022] Open
Abstract
Non-small-cell lung cancer (NSCLC) is a major health burden, and novel therapeutic options are needed to help solve this problem. One such option is immunotherapy, which targets immune checkpoint molecules that inhibit cancer cells, decreasing immune system activation, for example, immunotherapies target PD-1, its ligand PD-L1, and CTLA-4. There have been major advances in the development of agents that inhibit these molecules, called immune checkpoint inhibitors, and several of them are already approved for usage in NSCLC patients, especially in advanced stages. In this review, the reasons why immune checkpoint inhibitors could be beneficial and the clinical results of studies using these drugs for advanced or recurrent NSCLC patients are discussed, as is the safety profile of the drugs.
Collapse
Affiliation(s)
- Ratoe Suraya
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Motoko Tachihara
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
- Correspondence: Motoko Tachihara, Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan, Tel +81-78-382-5660, Fax +81-78-382-5661, Email
| | - Tatsuya Nagano
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yoshihiro Nishimura
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kazuyuki Kobayashi
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
22
|
Kasichayanula S, Mandlekar S, Shivva V, Patel M, Girish S. Evolution of Preclinical Characterization and Insights into Clinical Pharmacology of Checkpoint Inhibitors Approved for Cancer Immunotherapy. Clin Transl Sci 2022; 15:1818-1837. [PMID: 35588531 PMCID: PMC9372426 DOI: 10.1111/cts.13312] [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: 02/18/2022] [Revised: 05/05/2022] [Accepted: 05/09/2022] [Indexed: 11/30/2022] Open
Abstract
Cancer immunotherapy has significantly advanced the treatment paradigm in oncology, with approvals of immuno‐oncology agents for over 16 indications, many of them first line. Checkpoint inhibitors (CPIs) are recognized as an essential backbone for a successful anticancer therapy regimen. This review focuses on the US Food and Drug Administration (FDA) regulatory approvals of major CPIs and the evolution of translational advances since their first approval close to a decade ago. In addition, critical preclinical and clinical pharmacology considerations, an overview of the pharmacokinetic and dose/regimen aspects, and a discussion of the future of CPI translational and clinical pharmacology as combination therapy becomes a mainstay of industrial immunotherapy development and in clinical practice are also discussed.
Collapse
Affiliation(s)
| | | | - Vittal Shivva
- Genentech, 1 DNA Way, South San Francisco, 94080, CA
| | - Maulik Patel
- AbbVie Inc., 1000 Gateway Blvd, South San Francisco, 94080, CA
| | - Sandhya Girish
- Gilead Sciences, 310 Lakeside Drive, Foster City, 94404, CA
| |
Collapse
|
23
|
Araujo DV, Uchoa B, Soto-Castillo JJ, Furlan LL, Oliva M. When Less May Be Enough: Dose Selection Strategies for Immune Checkpoint Inhibitors Focusing on AntiPD-(L)1 Agents. Target Oncol 2022; 17:253-270. [PMID: 35687223 DOI: 10.1007/s11523-022-00890-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2022] [Indexed: 10/18/2022]
Abstract
Early clinical trials investigating antiPD(L)-1 agents rarely reached a maximum tolerated dose (MTD), and efficacy signals were observed even at the lowest dose levels. Most extended treatment intervals investigated indicated that these drugs do not follow a direct dose-toxicity or dose-efficacy relationship. Within this context and considering the high cost of antiPD(L)-1 agents, there is a significant debate on whether lower doses or the administration of such agents at an extended interval should be prospectively evaluated in already-approved agents, or at least be considered in novel combination trials involving antiPD(L)-1 drugs. Herein, we review the dosing, overall response rates, and incidence of treatment-related adverse events of antiPD(L)-1 agents in early dose-escalation trials and discuss the appropriateness of recommended Phase 2 dose selection as well as the final regulatory approved doses of such agents. Efficacy and safety data from randomized dose-range Phase 2 trials and real-world data (RWD) on the usage of lower doses and/or non-standard extended treatment intervals are also examined. As the accumulating evidence suggests lower doses or extended dosing intervals of antiPD(L)-1 may achieve a similar clinical benefit in comparison to the currently approved doses, we address the clinical and financial toxicity implications of using potentially higher doses than necessary. Last, we discuss ways to resolve the current dosing conundrum of antiPD-(L)1 agents such as performing near-equivalence studies and propose a framework for future development of immunotherapeutics to find the lowest efficacious dose instead of MTD.
Collapse
Affiliation(s)
- Daniel V Araujo
- Department of Medical Oncology, Hospital de Base/HB Onco, FUNFARME/FAMERP, Av. Brigadeiro Faria Lima 5544, São José do Rio Preto, SP, Brazil.
| | - Bruno Uchoa
- Department of Medical Oncology, Hospital de Base/HB Onco, FUNFARME/FAMERP, Av. Brigadeiro Faria Lima 5544, São José do Rio Preto, SP, Brazil
| | - Juan José Soto-Castillo
- Department of Medical Oncology, Institut Català d'Oncologia (ICO), L'Hospitalet de Llobregat, Av. Gran Via de L'Hospitalet 199-203, 08908, Barcelona, Spain
| | - Larissa L Furlan
- Department of Medical Oncology, Hospital de Base/HB Onco, FUNFARME/FAMERP, Av. Brigadeiro Faria Lima 5544, São José do Rio Preto, SP, Brazil
| | - Marc Oliva
- Department of Medical Oncology, Institut Català d'Oncologia (ICO), L'Hospitalet de Llobregat, Av. Gran Via de L'Hospitalet 199-203, 08908, Barcelona, Spain. .,Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
24
|
Challenges of the Immunotherapy: Perspectives and Limitations of the Immune Checkpoint Inhibitor Treatment. Int J Mol Sci 2022; 23:ijms23052847. [PMID: 35269988 PMCID: PMC8910928 DOI: 10.3390/ijms23052847] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/21/2022] [Accepted: 02/24/2022] [Indexed: 02/04/2023] Open
Abstract
Immunotherapy is a quickly developing type of treatment and the future of therapy in oncology. This paper is a review of recent findings in the field of immunotherapy with an emphasis on immune checkpoint inhibitors. The challenges that immunotherapy might face in near future, such as primary and acquired resistance and the irAEs, are described in this article, as well as the perspectives such as identification of environmental modifiers of immunity and development of anti-cancer vaccines and combined therapies. There are multiple factors that may be responsible for immunoresistance, such as genomic factors, factors related to the immune system cells or to the cancer microenvironment, factors emerging from the host cells, as well as other factors such as advanced age, biological sex, diet, many hormones, existing comorbidities, and the gut microbiome.
Collapse
|
25
|
Healey Bird B, Nally K, Ronan K, Clarke G, Amu S, Almeida AS, Flavin R, Finn S. Cancer Immunotherapy with Immune Checkpoint Inhibitors-Biomarkers of Response and Toxicity; Current Limitations and Future Promise. Diagnostics (Basel) 2022; 12:124. [PMID: 35054292 PMCID: PMC8775044 DOI: 10.3390/diagnostics12010124] [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: 12/23/2021] [Revised: 12/31/2021] [Accepted: 01/04/2022] [Indexed: 12/19/2022] Open
Abstract
Immune checkpoint inhibitors are monoclonal antibodies that are used to treat over one in three cancer patients. While they have changed the natural history of disease, prolonging life and preserving quality of life, they are highly active in less than 40% of patients, even in the most responsive malignancies such as melanoma, and cause significant autoimmune side effects. Licenced biomarkers include tumour Programmed Death Ligand 1 expression by immunohistochemistry, microsatellite instability, and tumour mutational burden, none of which are particularly sensitive or specific. Emerging tumour and immune tissue biomarkers such as novel immunohistochemistry scores, tumour, stromal and immune cell gene expression profiling, and liquid biomarkers such as systemic inflammatory markers, kynurenine/tryptophan ratio, circulating immune cells, cytokines and DNA are discussed in this review. We also examine the influence of the faecal microbiome on treatment outcome and its use as a biomarker of response and toxicity.
Collapse
Affiliation(s)
- Brian Healey Bird
- School of Medicine, University College Cork, T12 K8AF Cork, Ireland
- Bon Secours Hospital, T12 K8AF Cork, Ireland
| | - Ken Nally
- School of Biochemistry and Cell Biology, University College Cork, T12 K8AF Cork, Ireland;
- APC Microbiome Ireland, University College Cork, T12 K8AF Cork, Ireland; (G.C.); (A.S.A.)
| | - Karine Ronan
- Department of Oncology, St. Vincent’s University Hospital, D04 T6F4 Dublin, Ireland;
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, T12 K8AF Cork, Ireland; (G.C.); (A.S.A.)
- Department of Psychiatry, University College Cork, T12 K8AF Cork, Ireland
| | - Sylvie Amu
- Cancer Research at UCC, University College Cork, T12 K8AF Cork, Ireland;
| | - Ana S. Almeida
- APC Microbiome Ireland, University College Cork, T12 K8AF Cork, Ireland; (G.C.); (A.S.A.)
| | - Richard Flavin
- Department of Histopathology, Trinity College Dublin, D08 NHY1 Dublin, Ireland; (R.F.); (S.F.)
- St. James’s Hospital Dublin, D08 NHY1 Dublin, Ireland
| | - Stephen Finn
- Department of Histopathology, Trinity College Dublin, D08 NHY1 Dublin, Ireland; (R.F.); (S.F.)
- St. James’s Hospital Dublin, D08 NHY1 Dublin, Ireland
| |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW The resistance of immune checkpoint inhibitors (ICIs) has become an obstacle to further improve the survival of patients with advanced cancer. This review provides an overview of recent advances in primary resistance mechanisms of ICIs. RECENT FINDINGS With the improvement of study approach, new characteristics and trends have emerged in the classification of tumor immune subtypes. The effects of germline genetic on tumor microenvironment and the efficacy of immunotherapy have been further studied. Exosomal programmed death-ligand 1 (PD-L1) is an increasing focus of research in primary resistance mechanisms of ICIs. In addition to antibiotics and steroids, the influence of other concomitant medications on the efficacy of ICIs has recently gained more attention. SUMMARY Exploring the resistance mechanisms of ICIs is one of the great challenges in the field of tumor immunotherapy. Continued work to understand the resistance mechanism of ICIs is ongoing.
Collapse
Affiliation(s)
- Yi-Ze Li
- Department of Clinical Oncology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, PR China
| | | |
Collapse
|
27
|
Pesola G, Murianni V, Rebuzzi SE, Banna GL, Cerbone L, Catalano F, Borea R, Gandini A, Cremante M, Puglisi S, Trovato F, Fornarini G. Durable response after immunotherapy discontinuation for delayed and severe immune-related adverse events: a case report. Immunotherapy 2021; 13:1379-1386. [PMID: 34743545 DOI: 10.2217/imt-2021-0085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Recent studies have shown that immune-related adverse events (irAEs), occurring even after the discontinuation of immune checkpoint inhibitors (ICIs), may be associated with favorable disease outcomes, particularly in patients with melanoma and lung cancer. However, a few clinical cases have been described on the correlation between irAEs and ICIs efficacy in renal cell carcinoma (RCC) patients. This study reports the clinical case of a metastatic RCC patient who has experienced severe immune-related renal toxicity after 19 months of nivolumab use. Despite immunotherapy discontinuation, the patient has maintained clinical benefit and disease progression-free for 3 years. We examined the correlation between the occurrence and the severity of irAEs, treatment discontinuation and clinical benefits. The evidence on ICI retreatment following ICI discontinuation due to irAEs was also reviewed.
Collapse
Affiliation(s)
- Guido Pesola
- Clinic of Medical Oncology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Veronica Murianni
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, Genova, 16132, Italy
| | - Sara Elena Rebuzzi
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, Genova, 16132, Italy.,Department of Internal Medicine & Medical Specialties, University of Genova, Genova, 16132, Italy
| | - Giuseppe Luigi Banna
- Department of Oncology, Portsmouth Hospitals University NHS Trust, Portsmouth, PO6 3LY, UK
| | - Luigi Cerbone
- Département de médecine oncologique, Gustave Roussy, Villejuif, 94805, France
| | - Fabio Catalano
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, Genova, 16132, Italy
| | - Roberto Borea
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, Genova, 16132, Italy
| | - Annalice Gandini
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, Genova, 16132, Italy
| | - Malvina Cremante
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, Genova, 16132, Italy
| | - Silvia Puglisi
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, Genova, 16132, Italy
| | - Francesco Trovato
- Pharmacy Complex Unit, IRCCS Ospedale Policlinico San Martino, Genova, 16132, Italy
| | - Giuseppe Fornarini
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, Genova, 16132, Italy
| |
Collapse
|
28
|
Rebuzzi SE, Banna GL, Murianni V, Damassi A, Giunta EF, Fraggetta F, De Giorgi U, Cathomas R, Rescigno P, Brunelli M, Fornarini G. Prognostic and Predictive Factors in Advanced Urothelial Carcinoma Treated with Immune Checkpoint Inhibitors: A Review of the Current Evidence. Cancers (Basel) 2021; 13:5517. [PMID: 34771680 PMCID: PMC8583566 DOI: 10.3390/cancers13215517] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 10/26/2021] [Accepted: 11/01/2021] [Indexed: 12/13/2022] Open
Abstract
In recent years, the treatment landscape of urothelial carcinoma has significantly changed due to the introduction of immune checkpoint inhibitors (ICIs), which are the standard of care for second-line treatment and first-line platinum-ineligible patients with advanced disease. Despite the overall survival improvement, only a minority of patients benefit from this immunotherapy. Therefore, there is an unmet need to identify prognostic and predictive biomarkers or models to select patients who will benefit from ICIs, especially in view of novel therapeutic agents. This review describes the prognostic and predictive role, and clinical readiness, of clinical and tumour factors, including new molecular classes, tumour mutational burden, mutational signatures, circulating tumour DNA, programmed death-ligand 1, inflammatory indices and clinical characteristics for patients with urothelial cancer treated with ICIs. A classification of these factors according to the levels of evidence and grades of recommendation currently indicates both a prognostic and predictive value for ctDNA and a prognostic relevance only for concomitant medications and patients' characteristics.
Collapse
Affiliation(s)
- Sara Elena Rebuzzi
- Medical Oncology, Ospedale San Paolo, 17100 Savona, Italy
- Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genova, 16132 Genova, Italy
| | | | - Veronica Murianni
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (V.M.); (G.F.)
| | - Alessandra Damassi
- Academic Unit of Medical Oncology, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Emilio Francesco Giunta
- Department of Precision Medicine, Università Degli Studi della Campania Luigi Vanvitelli, 80131 Naples, Italy;
| | | | - Ugo De Giorgi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Richard Cathomas
- Division of Oncology/Hematology, Kantonsspital Graubünden, 7000 Chur, Switzerland;
| | - Pasquale Rescigno
- Interdisciplinary Group for Translational Research and Clinical Trials, Urogenital Cancers GIRT-Uro, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10060 Turin, Italy;
| | - Matteo Brunelli
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, 37134 Verona, Italy;
| | - Giuseppe Fornarini
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (V.M.); (G.F.)
| |
Collapse
|
29
|
Deligiorgi MV, Sagredou S, Vakkas L, Trafalis DT. The Continuum of Thyroid Disorders Related to Immune Checkpoint Inhibitors: Still Many Pending Queries. Cancers (Basel) 2021; 13:5277. [PMID: 34771441 PMCID: PMC8582503 DOI: 10.3390/cancers13215277] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/11/2021] [Accepted: 10/18/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Until more data are available to shed light on the thyroid disorders related to immune checkpoint inhibitors (ICPi) implemented for the treatment of hematological malignancies, the decision-making is guided by pertinent data derived mostly from solid tumors. METHODS The present review provides a comprehensive and updated overview of the thyroid disorders related to ICPi, namely to inhibitors of cytotoxic T-lymphocyte antigen 4 (CTLA-4), programmed cell death (PD) 1 (PD-1), and the ligand of the latter (PD-L1). RESULTS With the increasing recognition of ir thyroid disorders, many outstanding issues have emerged. Ir thyroid disorders are reminiscent of, but not identical to, thyroid autoimmunity. Interclass and intraclass ICPi differences regarding thyroid immunotoxicity await interpretation. The available data concerning the predictive value of thyroid autoantibodies for the development of ir thyroid disorders are inconclusive. Mounting data indicate an association of ir thyroid disorders with ICPi efficacy, but a causative link is still lacking. The path forward is a tailored approach, entailing: (i) the validation of tumor-specific, patient-specific, and ICPi-specific predictive factors; (ii) appropriate patient selection; (iii) the uncoupling of antitumor immunity from immunotoxicity; (iv) a multidisciplinary initiative; and (v) global registry strategies. CONCLUSIONS Untangling and harnessing the interrelationship of immuno-oncology with endocrinology underlying the ir thyroid disorders will yield the optimal patient care.
Collapse
Affiliation(s)
- Maria V. Deligiorgi
- Department of Pharmacology—Clinical Pharmacology Unit, Faculty of Medicine, National and Kapodistrian University of Athens, Building 16, 1st Floor, 75 Mikras Asias Str., Goudi, 11527 Athens, Greece; (S.S.); (L.V.); (D.T.T.)
| | | | | | | |
Collapse
|
30
|
Sanceau J, Gougelet A. Epigenetic mechanisms of liver tumor resistance to immunotherapy. World J Hepatol 2021; 13:979-1002. [PMID: 34630870 PMCID: PMC8473495 DOI: 10.4254/wjh.v13.i9.979] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/04/2021] [Accepted: 08/05/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver tumor, which stands fourth in rank of cancer-related deaths worldwide. The incidence of HCC is constantly increasing in correlation with the epidemic in diabetes and obesity, arguing for an urgent need for new treatments for this lethal cancer refractory to conventional treatments. HCC is the paradigm of inflammation-associated cancer, since more than 80% of HCC emerge consecutively to cirrhosis associated with a vast remodeling of liver microenvironment. In the recent decade, immunomodulatory drugs have been developed and have given impressive results in melanoma and later in several other cancers. In the present review, we will discuss the recent advancements concerning the use of immunotherapies in HCC, in particular those targeting immune checkpoints, used alone or in combination with other anti-cancers agents. We will address why these drugs demonstrate unsatisfactory results in a high proportion of liver cancers and the mechanisms of resistance developed by HCC to evade immune response with a focus on the epigenetic-related mechanisms.
Collapse
Affiliation(s)
- Julie Sanceau
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, Paris 75006, France
| | - Angélique Gougelet
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, Paris 75006, France
| |
Collapse
|
31
|
Kulikov A, Shipaeva E, Dmitrieva A, Batrak V, Shipunov G, Guy C, Smith J, Zhang R, Zhang M, Duan J, Chestukhin A, Barbashov S, Samsonov M, Lavrovsky Y. Preclinical Characterization of a Novel Anti-Cancer PD-L1 Inhibitor RPH-120. Front Pharmacol 2021; 12:723038. [PMID: 34456733 PMCID: PMC8386121 DOI: 10.3389/fphar.2021.723038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 07/26/2021] [Indexed: 12/02/2022] Open
Abstract
RPH-120 is a novel fully human anti-PD-L1 IgG1 monoclonal antibody with specifically designed Asn300Ala mutation in Fc fragment. Surface plasmon resonance assay showed that affinity of the RPH-120 to the dimeric form of human PD-L1-Fc fusion protein was much higher than affinity to the monomeric His-tagged PD-L1. Further binding studies demonstrated that RPH-120 is able to bind to human and monkey but not mouse PD-L1. Tissue cross-reactivity study showed good comparability of human and Cynomolgus monkeys tissue staining. Bioactivity was assessed using mixed lymphocyte reaction assay. This study revealed that RPH-120 was able to activate T cells preventing PD1/PD-L1 interaction. Antitumor efficacy was analyzed in HCC-827 lung cancer xenografts in humanized CD34+ mice at three dosage levels: 20, 80, and 200 mg/kg. RPH-120 demonstrated significant tumor growth inhibition, and this inhibition was comparable to that of atezolizumab. In a single dose toxicity, toxicokinetic and dose range finding study performed in Cynomolgus monkeys, RPH-120 was administered via intravenous (IV) bolus or 60-min IV infusion, followed by 8-weeks recovery period. An acceptable toxicokinetic profile was demonstrated and administration at doses of up to 200 mg/kg was well tolerated by all animals. In conclusion, RPH-120 revealed promising in vitro and in vivo activity and safety. RPH-120 is a potent anti-PD-L1 drug candidate for cancer immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Colin Guy
- Covance Laboratories Ltd, Harrogate, United Kingdom
| | - Jill Smith
- Covance Laboratories Ltd, Harrogate, United Kingdom
| | - Ran Zhang
- Covance Laboratories Ltd, Harrogate, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
32
|
Yeo MK, Sun P, Chung C, Park JH, Kang SH, Moon HS, Sung JK, Jeong HY, Kim JS. Clinical Significance of Composition Changes in T-cell Subpopulations After Chemotherapy in Patients With Gastric Cancer. In Vivo 2021; 35:2417-2424. [PMID: 34182525 DOI: 10.21873/invivo.12519] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/01/2021] [Accepted: 04/13/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND/AIM New therapeutic agents and prognostic biomarkers for gastric cancer are needed. We analyzed the composition of peripheral blood T-cell subpopulations in response to chemotherapy in patients with gastric cancer. PATIENTS AND METHODS Peripheral blood samples were collected from patients diagnosed with gastric cancer before and after chemotherapy (FOLFOX; oxaliplatin, 5-fluorouracil, and leucovorin). Peripheral blood mononuclear cells were isolated. Patients were divided into responder (n=5) and non-responder groups (n=2) based on their chemotherapy outcomes. RESULTS Non-responders showed lower numbers of CD4+/total cells and CD8+/total cells after chemotherapy compared to the responder group, but the difference was not significant (p=0.905, p=0.095). Naïve T, central memory T, effector memory T and effector T-cell counts differed in both groups after chemotherapy. CONCLUSION Changes in peripheral T-cell subpopulations after chemotherapy were confirmed in patients with gastric cancer, which may be a prognostic predictor and development of therapeutic agents.
Collapse
Affiliation(s)
- Min-Kyung Yeo
- Department of Pathology, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Pureum Sun
- Research Institute for Medical Sciences, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Chaeuk Chung
- Division of Pulmonology, Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Jae Ho Park
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Sun Hyung Kang
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Hee Seok Moon
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Jae Kyu Sung
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Hyun Yong Jeong
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Ju Seok Kim
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| |
Collapse
|
33
|
Brito ABC, Camandaroba MPG, de Lima VCC. Anti-PD1 versus anti-PD-L1 immunotherapy in first-line therapy for advanced non-small cell lung cancer: A systematic review and meta-analysis. Thorac Cancer 2021; 12:1058-1066. [PMID: 33586297 PMCID: PMC8017262 DOI: 10.1111/1759-7714.13867] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 01/12/2021] [Accepted: 01/12/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Due to the increasing number of trials with immune checkpoint inhibitors (ICIs) in the first-line therapy of non-small cell lung cancer (NSCLC) patients, we performed a systematic review and meta-analyses to investigate the difference between anti PD-1 and PD-L1 antibodies, used alone or in combination with chemotherapy, through adjusted indirect analysis to minimize the potential bias regarding overall survival (OS), progression-free survival (PFS), overall response rate (ORR) and grade 3-5 adverse events (AEs). METHODS A systematic review of studies reporting clinical outcomes and toxicity associated with first-line therapy employing anti-PD1 or anti-PD-L1 antibodies alone, or in combination with chemotherapy, to treat metastatic, treatment-naïve NSCLC patients was performed. Primary outcomes were OS, PFS, ORR and grade 3-5 AEs. We used a random-effects model to generate pooled estimates for proportions. Meta-analyses using pooled risk ratios were performed for binary outcomes from comparative studies with the random effects model. RESULTS A total of 13 eligible studies met our eligibility criteria, including 7673 patients. In the ICI-chemotherapy combination subgroup, we observed that anti-PD1 therapy was associated with better OS (p = 0.022) and PFS (p = 0.029) compared with anti-PD-L1 therapy. In the monotherapy subgroup, there was no statistical difference between the use of anti-PD-1 and anti-PD-L1 for OS and PFS. With regard to ORR and toxicity, in the ICI-chemotherapy combination subgroup, we observed a trend of better ORR (p = 0.12) with the use of anti-PD1 therapy and less frequent grade 3-5 AEs compared to the use of anti-PD-L1 therapy (p = 0.0302). In the monotherapy subgroup, there was no statistical difference between the use of anti-PD-1 and anti-PD-L1 regarding ORR and toxicity. CONCLUSIONS Our study suggests that PD-1 drug plus chemotherapy is superior to anti-PD-L1 plus chemotherapy for NSCLC; nevertheless, as monotherapy, both strategies appear to be similar.
Collapse
|
34
|
Landre T, Chouahnia K, Des Guetz G, Duchemann B, Assié JB, Chouaïd C. First-line immune-checkpoint inhibitor plus chemotherapy versus chemotherapy alone for extensive-stage small-cell lung cancer: a meta-analysis. Ther Adv Med Oncol 2020; 12:1758835920977137. [PMID: 33343721 PMCID: PMC7731693 DOI: 10.1177/1758835920977137] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/05/2020] [Indexed: 12/23/2022] Open
Abstract
Introduction Platin-based chemotherapy (CT) has long been the first-line standard-of-care for patients with extensive-stage small-cell lung cancer (ES-SCLC). Adding immune-checkpoint inhibitor(s) to CT (ICI+CT) in this setting is an option of interest, although its benefit is apparently modest. Methods This meta-analysis was conducted on randomized trials comparing first-line ICI+CT versus CT alone for ES-SCLC. Outcomes included overall survival (OS), progression-free survival (PFS), objective response rate (ORR), response at 12 months and adverse events (AEs). Subgroup analyses were computed according to the immunotherapy used, performance status (PS), age, platinum salt, liver metastases and brain metastases at diagnosis. Results The literature search identified one randomized phase II (ECOG-ACRIN-5161) and four phase III trials (CASPIAN, IMPOWER-133, KEYNOTE-604 and Reck et al. 2016) that included 2775 patients (66% males, 95% smokers, median age: 64 years, PS = 0 or 1). ICI+CT was significantly associated (hazard ratio [95% confidence interval]) with prolonged OS [0.82 (0.75-0.89); p < 0.00001] and PFS [0.81 (0.75-0.87); p < 0.00001], with OS benefits for anti-PD-L1 [0.73 (0.63-0.85); p < 0.0001] or anti-PD-1 [0.76 (0.63-0.93); p < 0.006] but not for anti-CTLA-4 [0.90 (0.80-1.01), p = 0.07]. ORRs for ICI+CT or CT alone were comparable [odds ratio 1.12 (0.97-1.00); p = 0.12], but responses at 12 months favored ICI+CT [4.16 (2.81-6.17), p < 0.00001]. Serious grade-3/4 AEs were more frequent with ICI+CT [odds ratio 1.18 (1.02-1.37); p = 0.03]. Compared with CT, no ICI+CT benefit was found for ES-SCLC with brain metastases at diagnosis [HR 1.14 (0.87-1.50); p = 0.34]. Conclusions First-line ICI+CT appears to be superior to CT alone for ES-SCLC except for patients with brain metastases at diagnosis.
Collapse
Affiliation(s)
- Thierry Landre
- Department of Public Health, HUPSSD, APHP, 125 Rue de Stalingrad, Bobigny, 93000, France
| | - Kader Chouahnia
- Service d'Oncologie, HUPSSD, APHP, Hôpital Avicenne, Bobigny, France
| | - Gaëtan Des Guetz
- Sevice d'Oncologie, Centre Hospitalier Delafontaine, Saint-Denis, France
| | - Boris Duchemann
- Service d'Oncologie, HUPSSD, APHP, Hôpital Avicenne, Bobigny, France
| | - Jean-Baptiste Assié
- Service de Pneumologie, Centre Hospitalier Intercommunal de Créteil, Créteil
| | - Christos Chouaïd
- Service de Pneumologie, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| |
Collapse
|
35
|
Ge X, Zhang Z, Zhang S, Yuan F, Zhang F, Yan X, Han X, Ma J, Wang L, Tao H, Li X, Zhi X, Huang Z, Hofman P, Prelaj A, Banna GL, Mutti L, Hu Y, Wang J. Immunotherapy beyond progression in patients with advanced non-small cell lung cancer. Transl Lung Cancer Res 2020; 9:2391-2400. [PMID: 33489801 PMCID: PMC7815351 DOI: 10.21037/tlcr-20-1252] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Background Immune checkpoint inhibitors (ICIs) represent a great breakthrough in the treatment of advanced non-small cell lung cancer (aNSCLC). However, whether immunotherapy beyond progression (IBP) is effective for aNSCLC has yet to be established. Therefore, a retrospective clinical study was conducted to investigate the efficacy of IBP in patients with aNSCLC under real-world conditions. Methods A total of 125 Chinese patients with aNSCLC who experienced progressive disease (PD) after receiving monotherapy or combination therapy (combined with chemotherapy or/and antiangiogenic therapy) with programmed cell death-1 (PD-1)/programmed cell death ligand-1 (PD-L1) inhibitors between January 2015 and March 2019 were enrolled. Patients who were treated with ICIs for more than 6 weeks after PD were defined as IBP (n=39), while those who received ICI treatment for less than 6 weeks or discontinued it due to PD were defined as non-IBP (n=86). Patient clinical characteristics were evaluated. An optimization-based method was applied to balance the clinical baseline characteristics between the two groups. Results In total population, the IBP group had longer overall survival (median OS, 26.6 vs. 9.5 months; HR, 0.40; 95% CI: 0.23–0.69; P<0.001) and progression-free survival (median PFS, 8.9 vs. 4.1 months; HR, 0.41; 95% CI: 0.26–0.65; P<0.001), compared with the non-IBP group. Despite no significant difference in objective response rate (ORR, 15.4% vs. 11.6%, P=0.560), disease control rate (DCR) was significantly higher in the IBP group (89.7% vs. 61.6%, P<0.001). After balancing baseline covariates, the IBP group also had longer OS (median: 26.6 vs. 10.7 months; HR, 0.40; 95% CI: 0.19–0.84; P=0.015) and PFS (median: 9.7 vs. 4.3 months; HR, 0.28; 95% CI: 0.15–0.51; P<0.001), with a benefit in either of patients previously treated with ICI monotherapy or in combination therapy and with non-response to the previously ICI. Conclusions IBP is associated with longer OS and PFS in patients with aNSCLC. Our findings may suggest new therapeutic options for patients with aNSCLC who experienced disease progression after initial immunotherapy.
Collapse
Affiliation(s)
- Xiangwei Ge
- Medical School of Chinese PLA, Beijing, China.,Department of Oncology, the Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhibo Zhang
- Department of Oncology, the Second Medical Center, Chinese PLA General Hospital, Beijing, China.,The 78th Group Army Hospital of Chinese PLA, Mudanjiang, China
| | - Sujie Zhang
- Department of Oncology, Chinese PLA General Hospital, Beijing, China
| | - Fang Yuan
- Department of Oncology, Chinese PLA General Hospital, Beijing, China
| | - Fan Zhang
- Department of Oncology, Chinese PLA General Hospital, Beijing, China
| | - Xiang Yan
- Department of Oncology, Chinese PLA General Hospital, Beijing, China
| | - Xiao Han
- Department of Oncology, Chinese PLA General Hospital, Beijing, China
| | - Junxun Ma
- Department of Oncology, Chinese PLA General Hospital, Beijing, China
| | - Lijie Wang
- Department of Oncology, Chinese PLA General Hospital, Beijing, China
| | - Haitao Tao
- Department of Oncology, Chinese PLA General Hospital, Beijing, China
| | - Xiaoyan Li
- Department of Oncology, Chinese PLA General Hospital, Beijing, China
| | - Xiaoyu Zhi
- Medical School of Chinese PLA, Beijing, China.,Department of Oncology, the Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | | | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Nice, France
| | - Arsela Prelaj
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy.,Department of Electronics, Information, and Bioengineering, Polytechnic University of Milan, Milan, Italy
| | - Giuseppe Luigi Banna
- Department of Oncology, Portsmouth Hospitals University NHS Trust, Portsmouth, UK
| | - Luciano Mutti
- Center for Biotechnology, Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, Philadelphia, PA, USA
| | - Yi Hu
- Department of Oncology, Chinese PLA General Hospital, Beijing, China
| | - Jinliang Wang
- Department of Oncology, the Second Medical Center, Chinese PLA General Hospital, Beijing, China.,Department of Oncology, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
36
|
Perrichet A, Ghiringhelli F, Rébé C. Understanding Inflammasomes and PD-1/PD-L1 Crosstalk to Improve Cancer Treatment Efficiency. Cancers (Basel) 2020; 12:cancers12123550. [PMID: 33261061 PMCID: PMC7761387 DOI: 10.3390/cancers12123550] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 11/25/2020] [Accepted: 11/25/2020] [Indexed: 12/20/2022] Open
Abstract
Inflammasomes and immune checkpoints have been shown to participate in carcinogenesis, cancer growth and response to treatment. Thus, targeting cytokines resulting from inflammasome activation, such as interleukin (IL)-1β, has emerged as a new tool in the therapeutic arsenal. Moreover, the use of checkpoint inhibitors such as anti-PD-1 or anti-PD-L1 has revolutionized the treatment of some cancer patients. However, inflammasome activation and consecutive cytokine release only occurs in some chemotherapeutic treatments and immune checkpoint inhibitors only work for a restricted number of patients, thus limiting the use of therapies targeting these pathways. Expanding knowledge about the inefficiency of these therapies recently brought forward the hypothesis of targeting both pathways. In this review, we provide an overview of the crosstalk between inflammasomes and programmed death-1 (PD-1)/programmed death-ligand 1 (PD-L1) that might explain how these two pathways are mutually dependent, and perhaps why targeting only one of them leads to inefficiency of cancer treatment in some patients.
Collapse
Affiliation(s)
| | | | - Cédric Rébé
- Correspondence: ; Tel.: +33-(0)3-80-73-77-90
| |
Collapse
|