1
|
Courtney PT, Venkat PS, Shih YCT, Chang AJ, Lee A, Steinberg ML, Raldow AC. Cost-Effectiveness of Pembrolizumab With Chemoradiotherapy for Locally Advanced Cervical Cancer. JAMA Netw Open 2025; 8:e250033. [PMID: 40036034 PMCID: PMC11880949 DOI: 10.1001/jamanetworkopen.2025.0033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 12/19/2024] [Indexed: 03/06/2025] Open
Abstract
Importance The KEYNOTE-A18 trial demonstrated that adding concurrent and adjuvant pembrolizumab to chemoradiotherapy and brachytherapy significantly improved survival in patients with newly diagnosed, locally advanced cervical cancer. However, considering the annual global incidence of 660 000 cases of cervical cancer, including 13 820 in the US in 2024, incorporating this regimen into the standard of care could have substantial health care economic implications for both patients and the health care system. Objective To determine the cost-effectiveness of adding pembrolizumab to the first-line treatment of newly diagnosed, locally advanced cervical cancer. Design, Setting, and Participants This economic evaluation created a Markov model simulating 50-year outcomes to evaluate cost-effectiveness from the payer perspective for patients receiving either pembrolizumab or placebo in addition to chemoradiotherapy plus brachytherapy. Probabilities, including disease progression, survival, and treatment-related toxic effects, were derived from KEYNOTE-A18 clinical trial data in patients with newly diagnosed, locally advanced cervical cancer. Costs and health utilities were obtained from published literature; 1-way, 3-way, and probabilistic sensitivity analyses were used to assess model uncertainty. Data analyses were conducted from April to November 2024. Exposure Pembrolizumab. Main Outcomes and Measures Costs, measured in 2024 US dollars, and effectiveness, measured in quality-adjusted life-years (QALYs) were used to calculate an incremental cost-effectiveness ratio (ICER). A willingness-to-pay threshold of $100 000 per QALY was chosen, below which pembrolizumab would be considered cost-effective. Results KEYNOTE-A18 enrolled 1060 patients (529 in pembrolizumab group, 531 in placebo group). The median age was 50 years. Pembrolizumab increased costs by $257 000 and effectiveness by 1.40 QALYs, yielding an incremental cost-effectiveness ratio of $183 400 per QALY. The addition of pembrolizumab became cost-effective if its monthly cost was decreased from $16 990 to $9190 (a 45.6% reduction) or its maximum duration of 24 months was decreased to 10 months. The model was insensitive to assumptions about treatment-related toxic effects, progression-free survival, and overall survival. Probabilistic sensitivity analysis indicated that at a willingness-to-pay threshold of $100 000 per QALY, the addition of pembrolizumab was cost-effective 37.3% of the time. Conclusions and Relevance In this economic evaluation of adding concurrent and adjuvant pembrolizumab to first-line treatment of newly diagnosed, locally advanced cervical cancer, this regimen was not cost-effective at current prices despite data demonstrating improved survival with this regimen.
Collapse
Affiliation(s)
| | - Puja S. Venkat
- Department of Radiation Oncology, University of California, Los Angeles
| | - Ya-Chen Tina Shih
- Department of Radiation Oncology, University of California, Los Angeles
| | - Albert J. Chang
- Department of Radiation Oncology, University of California, Los Angeles
| | - Alan Lee
- Department of Radiation Oncology, University of California, Los Angeles
| | | | - Ann C. Raldow
- Department of Radiation Oncology, University of California, Los Angeles
| |
Collapse
|
2
|
Lee SSY, Pagacz J, Averbek S, Scholten D, Liu Y, Kron SJ. Timing Anti-PD-L1 Checkpoint Blockade Immunotherapy to Enhance Tumor Irradiation. Cancers (Basel) 2025; 17:391. [PMID: 39941761 PMCID: PMC11815760 DOI: 10.3390/cancers17030391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/14/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Background: The ability of radiotherapy (RT) to drive anti-tumor immunity is limited by adaptive resistance. While RT induces inflammation and recruits activated tumor-infiltrating lymphocytes (TILs), including cytotoxic T lymphocytes (CTLs), the resulting radiation- and IFNγ-dependent PD-L1 expression restores an immunosuppressed tumor microenvironment. Unleashing an effective anti-tumor response may require the precise sequencing of RT and checkpoint blockade immunotherapy (CBI) to block PD-L1 signaling before it can mediate its suppressive effects. Methods: Flank tumors formed in BALB/c mice with syngeneic CT26 colon or 4T1 mammary carcinoma cells were treated with otherwise ineffective doses of ionizing radiation (10 Gy) followed by CBI (0.2 mg anti-PD-L1, i.v.) after 0, 1, 3, 5, or 7 days, comparing tumor response. Anti-PD-L1 delivery was measured by fluorescence, TILs by flow cytometry and immunofluorescence, PD-L1 expression by immunohistochemistry, and tumor size by calipers. Results: In both CT26 and 4T1 tumors, 10 Gy alone resulted in a transient growth delay associated with infiltrating CTLs peaking at 3 days and PD-L1 at 5 days. CTLs returned to baseline after 7 days, consistent with adaptive resistance. Anti-PD-L1 failed to potentiate radiation except when injected 5 days after 10 Gy, which prevented CTL depletion and led to tumor elimination. Potentially contributing to compound effects, anti-PD-L1 penetrated tumors and bound PD-L1 more efficiently after irradiation. Conclusions: Optimal timing to exploit radiation-induced permeability to enhance CBI delivery and interrupt adaptive resistance by blocking PD-L1 as it peaks may offer a general strategy to enhance external beam radiotherapy by protecting activated TILs and potentiating anti-tumor immune response.
Collapse
Affiliation(s)
- Steve Seung-Young Lee
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Joanna Pagacz
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA; (J.P.); (S.A.); (D.S.); (Y.L.)
| | - Sera Averbek
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA; (J.P.); (S.A.); (D.S.); (Y.L.)
| | - David Scholten
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA; (J.P.); (S.A.); (D.S.); (Y.L.)
| | - Yue Liu
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA; (J.P.); (S.A.); (D.S.); (Y.L.)
| | - Stephen J. Kron
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA; (J.P.); (S.A.); (D.S.); (Y.L.)
| |
Collapse
|
3
|
Elghawy O, Barsouk A, Patel R, Reed-Guy L, Wang JS, Xu J, Yang A, Sussman J, Kaur V. Impact of immune-related adverse events on survival among patients with head-and-neck squamous cell carcinoma. Immunotherapy 2024; 16:1069-1078. [PMID: 39392156 PMCID: PMC11492630 DOI: 10.1080/1750743x.2024.2409617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 09/24/2024] [Indexed: 10/12/2024] Open
Abstract
Aim: Immune-checkpoint inhibitors (ICIs) have revolutionized treatment of metastatic head and neck squamous cell carcinomas (HNSCCs). Our goal was to assess for an association between immune-related adverse events (irAEs) and clinical outcomes for patients on ICIs.Methods: We analyzed a cohort of 110 HNSCC patients who received ICI therapy at the University of Virginia.Results: On review, 48% of our patients experienced an irAE with the most common events being hypothyroidism (30%), dermatitis (14%) and hepatitis (11%). Women were more likely to experience irAEs. Treatment interruption/discontinuation occurred in 43% patients with irAEs. Development of irAEs was associated with superior objective response rate (68 vs. 39%, p = 0.009), with a greater rate of CR (17 vs. 5%) and PR (32 vs. 16%). Twelve patients underwent ICI re-treatment following irAE, with 17% attaining a complete disease response, 25% attaining a partial response, 33% achieving stable disease and 25% experiencing disease progression with ICI resumption.Conclusion: Development of irAE was associated with superior objective response rate, with a greater rate of CR and PR. ICI re-treatment following irAE was feasible in a significant proportion of patients and can be attempted in carefully selected patients, given the dearth of second-line therapies for these patients.
Collapse
Affiliation(s)
- Omar Elghawy
- Hospital of the University of Pennsylvania, Department of Internal Medicine, Philadelphia, PA19104, USA
| | - Adam Barsouk
- Hospital of the University of Pennsylvania, Department of Internal Medicine, Philadelphia, PA19104, USA
| | - Reema Patel
- University of Virginia Health, Department of Internal Medicine, Division of Hematology & Oncology, Charlottesville, VA22903, USA
| | - Lauren Reed-Guy
- Hospital of the University of Pennsylvania, Department of Internal Medicine, Philadelphia, PA19104, USA
| | - John S Wang
- McGaw Medical Center of Northwestern University, Department of Internal Medicine, Chicago, IL60611, USA
| | - Jessica Xu
- Hospital of the University of Pennsylvania, Department of Internal Medicine, Philadelphia, PA19104, USA
| | - Austin Yang
- Hospital of the University of Pennsylvania, Department of Internal Medicine, Philadelphia, PA19104, USA
| | - Jonathan Sussman
- Hospital of the University of Pennsylvania, Department of Internal Medicine, Philadelphia, PA19104, USA
| | - Varinder Kaur
- University of Virginia Health, Department of Internal Medicine, Division of Hematology & Oncology, Charlottesville, VA22903, USA
| |
Collapse
|
4
|
Srivastava S, Tyagi A, Pawar VA, Khan NH, Arora K, Verma C, Kumar V. Revolutionizing Immunotherapy: Unveiling New Horizons, Confronting Challenges, and Navigating Therapeutic Frontiers in CAR-T Cell-Based Gene Therapies. Immunotargets Ther 2024; 13:413-433. [PMID: 39219644 PMCID: PMC11365499 DOI: 10.2147/itt.s474659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
The CAR-T cell therapy has marked the dawn of new era in the cancer therapeutics and cell engineering techniques. The review emphasizes on the challenges that obstruct the therapeutic efficiency caused by cell toxicities, immunosuppressive tumor environment, and decreased T cell infiltration. In the interest of achieving the overall survival (OS) and event-free survival (EFS) of patients, the conceptual background of potential target selection and various CAR-T cell design techniques are described which can minimize the off-target effects, reduce toxicity, and thus increase the resilience of CAR-T cell treatment in the haematological malignancies as well as in solid tumors. Furthermore, it delves into cutting-edge technologies like gene editing and synthetic biology, providing new opportunities to enhance the functionality of CAR-T cells and overcome mechanisms of immune evasion. This review provides a comprehensive understanding of the complex and diverse aspects of CAR-T cell-based gene treatments, including both scientific and clinical aspects. By effectively addressing the obstacles and utilizing the capabilities of cutting-edge technology, CAR-T cell therapy shows potential in fundamentally changing immunotherapy and reshaping the approach to cancer treatment.
Collapse
Affiliation(s)
- Shivani Srivastava
- Department of Pathology, School of Medicine, Yale University, New Haven, CT, USA
| | - Anuradha Tyagi
- Department of cBRN, Institute of Nuclear Medicine and Allied Science, Delhi, India
| | | | - Nawaid Hussain Khan
- Faculty of Medicine, Ala-Too International University, Bishkek, Kyrgyz Republic
| | - Kavita Arora
- Advanced Instrumentation Research Facility, Jawaharlal Nehru University, New Delhi, India
- School of Computational & Integrative Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Chaitenya Verma
- Department of Pathology, Wexner Medical Center, Ohio State University, Columbus, OH, USA
- Department of Biotechnology, SSET, Sharda University, Greater Noida, 201306, India
| | - Vinay Kumar
- Pennsylvania State University Hershey Medical Center, 500 University Dr, Heshey, PA, USA
| |
Collapse
|
5
|
Coussement M, Fazio R, Audisio A, El Khoury R, Abbassi FZ, Assaf I, Conti C, Gallio C, Benhima N, Bregni G, Gkolfakis P, Spagnolo V, Anthoine G, Liberale G, Moretti L, Martinive P, Hendlisz A, Demetter P, Sclafani F. PD-L1 Expression in Paired Samples of Rectal Cancer. Cancers (Basel) 2024; 16:2606. [PMID: 39061244 PMCID: PMC11275196 DOI: 10.3390/cancers16142606] [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: 06/16/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Immune checkpoint inhibitors and immune-related biomarkers are increasingly investigated in rectal cancer (RC). We retrospectively analysed PD-L1 expression in diagnostic biopsy and resection samples from RC patients treated at our centre between 2000 and 2020. PD-L1 immunostaining (22C3 clone) was evaluated according to tumour proportion (TPS), immune cell (ICS), and the combined positive score (CPS). Eighty-three patients were included. At diagnosis, PD-L1 expression ≥1%/≥5% was observed in 15.4%/0%, 80.7%/37.4%, and 69.2%/25.6% of patients based on TPS, ICS, and CPS, respectively. At surgery, the respective figures were 4.6%/1.5%, 60.2%/32.5%, and 50.7%/26.2%. Using the 1% cut-off and regardless of the scoring system, PD-L1 was less expressed in surgery than biopsy samples (p ≤ 0.04). In paired specimens, PD-L1-ICS reduction was especially observed following neoadjuvant long-course (chemo)radiotherapy (p = 0.03). PD-L1-ICS of ≥5% in surgical samples (HR: 0.17; p = 0.02), and a biopsy-to-surgery increase in PD-L1-ICS (HR: 0.19; p = 0.04) was predictive for longer disease-free survival, while the PD-L1-ICS of either ≥1% (HR 0.28; p = 0.04) or ≥5% (HR 0.19; p = 0.03) in surgical samples and the biopsy-to-surgery increase in PD-L1-ICS (HR: 0.20; p = 0.04) were associated with better overall survival. Our study suggests that PD-L1 expression in RC is largely reflective of immune cell infiltration, and its presence/increase in surgical samples predicts better outcomes.
Collapse
Affiliation(s)
- Mina Coussement
- Institut Jules Bordet, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), 1070 Brussels, Belgium; (M.C.); (R.F.); (V.S.)
| | - Roberta Fazio
- Institut Jules Bordet, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), 1070 Brussels, Belgium; (M.C.); (R.F.); (V.S.)
| | - Alessandro Audisio
- Institut Jules Bordet, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), 1070 Brussels, Belgium; (M.C.); (R.F.); (V.S.)
| | - Reem El Khoury
- Institut Jules Bordet, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), 1070 Brussels, Belgium; (M.C.); (R.F.); (V.S.)
| | - Fatima-Zahra Abbassi
- Institut Jules Bordet, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), 1070 Brussels, Belgium; (M.C.); (R.F.); (V.S.)
| | - Irene Assaf
- Institut Jules Bordet, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), 1070 Brussels, Belgium; (M.C.); (R.F.); (V.S.)
| | - Chiara Conti
- Institut Jules Bordet, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), 1070 Brussels, Belgium; (M.C.); (R.F.); (V.S.)
| | - Chiara Gallio
- Institut Jules Bordet, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), 1070 Brussels, Belgium; (M.C.); (R.F.); (V.S.)
| | - Nada Benhima
- Institut Jules Bordet, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), 1070 Brussels, Belgium; (M.C.); (R.F.); (V.S.)
| | - Giacomo Bregni
- Institut Jules Bordet, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), 1070 Brussels, Belgium; (M.C.); (R.F.); (V.S.)
| | - Paraskevas Gkolfakis
- Institut Jules Bordet, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), 1070 Brussels, Belgium; (M.C.); (R.F.); (V.S.)
| | - Valentina Spagnolo
- Institut Jules Bordet, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), 1070 Brussels, Belgium; (M.C.); (R.F.); (V.S.)
| | - Geraldine Anthoine
- Institut Jules Bordet, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), 1070 Brussels, Belgium; (M.C.); (R.F.); (V.S.)
| | - Gabriel Liberale
- Institut Jules Bordet, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), 1070 Brussels, Belgium; (M.C.); (R.F.); (V.S.)
| | - Luigi Moretti
- Institut Jules Bordet, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), 1070 Brussels, Belgium; (M.C.); (R.F.); (V.S.)
| | - Philippe Martinive
- Institut Jules Bordet, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), 1070 Brussels, Belgium; (M.C.); (R.F.); (V.S.)
| | - Alain Hendlisz
- Institut Jules Bordet, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), 1070 Brussels, Belgium; (M.C.); (R.F.); (V.S.)
| | - Pieter Demetter
- Cerba Path, Division CMP, 1070 Brussels, Belgium
- Laboratory for Experimental Gastroenterology, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Francesco Sclafani
- Institut Jules Bordet, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), 1070 Brussels, Belgium; (M.C.); (R.F.); (V.S.)
| |
Collapse
|
6
|
Fazio R, Audisio A, Daprà V, Conti C, Benhima N, Abbassi FZ, Assaf I, Hendlisz A, Sclafani F. Non-operative management after immune checkpoint inhibitors for early-stage, dMMR/MSI-H gastrointestinal cancers. Cancer Treat Rev 2024; 128:102752. [PMID: 38772170 DOI: 10.1016/j.ctrv.2024.102752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/10/2024] [Accepted: 05/11/2024] [Indexed: 05/23/2024]
Abstract
Surgery is a standard treatment for early-stage gastrointestinal cancers, often preceded by neoadjuvant chemo(radio)therapy or followed by adjuvant therapy. While leading to cure in a proportion of patients, it has some drawbacks such as intra/post-operative complications, mutilation and life-long functional sequelae. Further to the unprecedented efficacy data from studies of immune checkpoint inhibitors for advanced mismatch repair deficient/microsatellite instable (dMMR/MSI-H) tumours, a strong interest has recently emerged for the investigation of such agents in the neoadjuvant setting. Although limited by the exploratory design and small sample size, trials of neoadjuvant immune checkpoint inhibitors for early-stage dMMR/MSI-H gastrointestinal cancers have consistently reported complete response rates ranging from 70 % to 100 %. As a result, the question has arisen as to whether surgery is still needed or organ-preserving strategies should be offered to this especially immuno-sensitive population. In this article, we discuss the available evidence for neoadjuvant immune checkpoint inhibitors in dMMR/MSI-H gastrointestinal cancers and analyse opportunities and challenges to the implementation of non-operative management approaches in this setting.
Collapse
Affiliation(s)
- Roberta Fazio
- Université libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels, Belgium
| | - Alessandro Audisio
- Université libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels, Belgium
| | - Valentina Daprà
- Université libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels, Belgium
| | - Chiara Conti
- Université libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels, Belgium
| | - Nada Benhima
- Université libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels, Belgium
| | - Fatima-Zahara Abbassi
- Université libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels, Belgium
| | - Irene Assaf
- Université libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels, Belgium
| | - Alain Hendlisz
- Université libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels, Belgium
| | - Francesco Sclafani
- Université libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels, Belgium.
| |
Collapse
|
7
|
Telarovic I, Yong CSM, Kurz L, Vetrugno I, Reichl S, Fernandez AS, Cheng HW, Winkler R, Guckenberger M, Kipar A, Ludewig B, Pruschy M. Delayed tumor-draining lymph node irradiation preserves the efficacy of combined radiotherapy and immune checkpoint blockade in models of metastatic disease. Nat Commun 2024; 15:5500. [PMID: 38951172 PMCID: PMC11217506 DOI: 10.1038/s41467-024-49873-y] [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: 08/07/2023] [Accepted: 06/21/2024] [Indexed: 07/03/2024] Open
Abstract
Cancer resistance to immune checkpoint inhibitors motivated investigations into leveraging the immunostimulatory properties of radiotherapy to overcome immune evasion and to improve treatment response. However, clinical benefits of radiotherapy-immunotherapy combinations have been modest. Routine concomitant tumor-draining lymph node irradiation (DLN IR) might be the culprit. As crucial sites for generating anti-tumor immunity, DLNs are indispensable for the in situ vaccination effect of radiotherapy. Simultaneously, DLN sparing is often not feasible due to metastatic spread. Using murine models of metastatic disease in female mice, here we demonstrate that delayed (adjuvant), but not neoadjuvant, DLN IR overcomes the detrimental effect of concomitant DLN IR on the efficacy of radio-immunotherapy. Moreover, we identify IR-induced disruption of the CCR7-CCL19/CCL21 homing axis as a key mechanism for the detrimental effect of DLN IR. Our study proposes delayed DLN IR as a strategy to maximize the efficacy of radio-immunotherapy across different tumor types and disease stages.
Collapse
Affiliation(s)
- Irma Telarovic
- Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Carmen S M Yong
- Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Immunology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Lisa Kurz
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Irene Vetrugno
- Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Sabrina Reichl
- Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Alba Sanchez Fernandez
- Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Hung-Wei Cheng
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Rona Winkler
- Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Matthias Guckenberger
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Anja Kipar
- Laboratory for Animal Model Pathology, Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Burkhard Ludewig
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Martin Pruschy
- Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
8
|
Durante M. Kaplan lecture 2023: lymphopenia in particle therapy. Int J Radiat Biol 2024; 100:669-677. [PMID: 38442137 DOI: 10.1080/09553002.2024.2324472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 02/02/2024] [Indexed: 03/07/2024]
Abstract
PURPOSE Lymphopenia is now generally recognized as a negative prognostic factor in radiotherapy. Already at the beginning of the century we demonstrated that high-energy carbon ions induce less damage to the lymphocytes of radiotherapy patients than X-rays, even if heavy ions are more effective per unit dose in the induction of chromosomal aberrations in blood cells irradiated ex-vivo. The explanation was based on the volume effect, i.e. the sparing of larger volumes of normal tissue in Bragg peak therapy. Here we will review the current knowledge about the difference in lymphopenia between particle and photon therapy and the consequences. CONCLUSIONS There is nowadays an overwhelming evidence that particle therapy reduces significantly the radiotherapy-induced lymphopenia in several tumor sites. Because lymphopenia turns down the immune response to checkpoint inhibitors, it can be predicted that particle therapy may be the ideal partner for combined radiation and immunotherapy treatment and should be selected for patients where severe lymphopenia is expected after X-rays.
Collapse
Affiliation(s)
- Marco Durante
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
- Institute for Condensed Matter Physics, Technische Universität Darmstadt, Darmstadt, Germany
- Dipartimento di Fisica "Ettore Pancini", Università Federico II, Naples, Italy
| |
Collapse
|
9
|
Verma N, Renauer PA, Dong C, Xin S, Lin Q, Zhang F, Glazer PM, Chen S. Genome scale CRISPR screens identify actin capping proteins as key modulators of therapeutic responses to radiation and immunotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.14.575614. [PMID: 38293095 PMCID: PMC10827061 DOI: 10.1101/2024.01.14.575614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Radiotherapy (RT), is a fundamental treatment for malignant tumors and is used in over half of cancer patients. As radiation can promote anti-tumor immune effects, a promising therapeutic strategy is to combine radiation with immune checkpoint inhibitors (ICIs). However, the genetic determinants that impact therapeutic response in the context of combination therapy with radiation and ICI have not been systematically investigated. To unbiasedly identify the tumor intrinsic genetic factors governing such responses, we perform a set of genome-scale CRISPR screens in melanoma cells for cancer survival in response to low-dose genotoxic radiation treatment, in the context of CD8 T cell co-culture and with anti-PD1 checkpoint blockade antibody. Two actin capping proteins, Capza3 and Capg, emerge as top hits that upon inactivation promote the survival of melanoma cells in such settings. Capza3 and Capg knockouts (KOs) in mouse and human cancer cells display persistent DNA damage due to impaired homology directed repair (HDR); along with increased radiation, chemotherapy, and DNA repair inhibitor sensitivity. However, when cancer cells with these genes inactivated were exposed to sublethal radiation, inactivation of such actin capping protein promotes activation of the STING pathway, induction of inhibitory CEACAM1 ligand expression and resistance to CD8 T cell killing. Patient cancer genomics analysis reveals an increased mutational burden in patients with inactivating mutations in CAPG and/or CAPZA3, at levels comparable to other HDR associated genes. There is also a positive correlation between CAPG expression and activation of immune related pathways and CD8 T cell tumor infiltration. Our results unveil the critical roles of actin binding proteins for efficient HDR within cancer cells and demonstrate a previously unrecognized regulatory mechanism of therapeutic response to radiation and immunotherapy.
Collapse
Affiliation(s)
- Nipun Verma
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- System Biology Institute, Yale University, West Haven, Connecticut, USA
- Department of Therapeutic Radiology, Yale University, New Haven, Connecticut, USA
| | - Paul A. Renauer
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- System Biology Institute, Yale University, West Haven, Connecticut, USA
| | - Chuanpeng Dong
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- System Biology Institute, Yale University, West Haven, Connecticut, USA
| | - Shan Xin
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- System Biology Institute, Yale University, West Haven, Connecticut, USA
| | - Qianqian Lin
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- System Biology Institute, Yale University, West Haven, Connecticut, USA
| | - Feifei Zhang
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- System Biology Institute, Yale University, West Haven, Connecticut, USA
| | - Peter M. Glazer
- Department of Therapeutic Radiology, Yale University, New Haven, Connecticut, USA
- Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sidi Chen
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
- System Biology Institute, Yale University, West Haven, Connecticut, USA
- Immunobiology Program, Yale University, New Haven, Connecticut, USA
- Molecular Cell Biology, Genetics, and Development Program, Yale University, New Haven, Connecticut, USA
- Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Yale Stem Cell Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Yale Center for Biomedical Data Science, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
10
|
Belaidi L, Wang P, Quintin K, Durdux C, Giroux-Leprieur E, Giraud P. Impact of Waiting Response Evaluation to First-Line Systemic Therapy before Considering Local Ablative Therapy in Metastatic Non-Small-Cell Lung Cancer. Cancers (Basel) 2023; 15:5127. [PMID: 37958302 PMCID: PMC10649273 DOI: 10.3390/cancers15215127] [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: 09/17/2023] [Revised: 10/21/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
Stereotactic radiotherapy (SRT) is gaining increasing importance in metastatic non-small-cell lung cancer (mNSCLC) management. The optimal sequence of tumor irradiation relative to systemic treatment remains unclear. If waiting response evaluation to first-line systemic therapy (FLST) before considering local treatment may allow for the exclusion of poorer prognosis progressive tumors that may not benefit from SRT, performing irradiation near immune check point inhibitor (ICI) first administration seems to improve their synergic effect. Herein, we aimed to determine whether delaying SRT after response evaluation to FLST would result in better prognosis. We compared overall survival (OS), progression-free survival (PFS), and time to first subsequent therapy (TFST) for 50 patients locally treated before or within 90 days of initiating FLST (early SRT), with 49 patients treated at least 90 days after initiating FLST (late SRT). Patients treated with conventional chemotherapy alone exhibited significantly poorer median OS, PFS, and TFST in the early SRT arm: (in months) 16.5 [8.33-NR] vs. 58.3 [35.05-NR] (p = 0.0015); 4.69 [3.57-8.98] vs. 8.20 [6.66-12.00] (p = 0.017); and 6.26 [4.82-11.8] vs. 10.0 [7.44-21.8] (p = 0.0074), respectively. Patient receiving ICI showed no difference in OS (NR [25.2-NR] vs. 36.6 [35.1-NR], p = 0.79), PFS (7.54 [6.23-NR] vs. 4.07 [2.52-NR], p = 0.19), and TFST (13.7 [9.48-NR] vs. 10.3 [3.54-NR], p = 0.49). These results suggest that delaying SRT treatment in order to filter a rapidly growing tumor may be less necessary when ICI is administered in mNSCLC.
Collapse
Affiliation(s)
- Lahcene Belaidi
- Department of Radiation Oncology, Hôpital Européen Georges Pompidou AP-HP, 20 Rue Leblanc, 75015 Paris, France
| | - Pascal Wang
- Department of Pulmonology and Thoracic Oncology Service, Hôpital Ambroise Paré, 9 Av. Charles de Gaulle, 92100 Boulogne-Billancourt, France
| | - Kevin Quintin
- Department of Radiation Oncology, Hôpital Européen Georges Pompidou AP-HP, 20 Rue Leblanc, 75015 Paris, France
| | - Catherine Durdux
- Department of Radiation Oncology, Hôpital Européen Georges Pompidou AP-HP, 20 Rue Leblanc, 75015 Paris, France
| | - Etienne Giroux-Leprieur
- Department of Pulmonology and Thoracic Oncology Service, Hôpital Ambroise Paré, 9 Av. Charles de Gaulle, 92100 Boulogne-Billancourt, France
| | - Philippe Giraud
- Department of Radiation Oncology, Hôpital Européen Georges Pompidou AP-HP, 20 Rue Leblanc, 75015 Paris, France
| |
Collapse
|
11
|
Echavidre W, Fagret D, Faraggi M, Picco V, Montemagno C. Recent Pre-Clinical Advancements in Nuclear Medicine: Pioneering the Path to a Limitless Future. Cancers (Basel) 2023; 15:4839. [PMID: 37835533 PMCID: PMC10572076 DOI: 10.3390/cancers15194839] [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: 08/21/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 10/15/2023] Open
Abstract
The theranostic approach in oncology holds significant importance in personalized medicine and stands as an exciting field of molecular medicine. Significant achievements have been made in this field in recent decades, particularly in treating neuroendocrine tumors using 177-Lu-radiolabeled somatostatin analogs and, more recently, in addressing prostate cancer through prostate-specific-membrane-antigen targeted radionuclide therapy. The promising clinical results obtained in these indications paved the way for the further development of this approach. With the continuous discovery of new molecular players in tumorigenesis, the development of novel radiopharmaceuticals, and the potential combination of theranostics agents with immunotherapy, nuclear medicine is poised for significant advancements. The strategy of theranostics in oncology can be categorized into (1) repurposing nuclear medicine agents for other indications, (2) improving existing radiopharmaceuticals, and (3) developing new theranostics agents for tumor-specific antigens. In this review, we provide an overview of theranostic development and shed light on its potential integration into combined treatment strategies.
Collapse
Affiliation(s)
- William Echavidre
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (W.E.); (V.P.)
| | - Daniel Fagret
- Laboratory of Bioclinical Radiopharmaceutics, Universite Grenoble Alpes, CHU Grenoble Alpes, Inserm, 38000 Grenoble, France;
| | - Marc Faraggi
- Nuclear Medicine Department, Centre Hospitalier Princesse Grace, 98000 Monaco, Monaco;
| | - Vincent Picco
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (W.E.); (V.P.)
| | - Christopher Montemagno
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (W.E.); (V.P.)
| |
Collapse
|
12
|
Zhao XH, Gao HM, Wen JY, Wang HS, Wu LY, Song CY, Deng WZ, Zhu SC, Shen WB. Immune checkpoint inhibitors combined with or without radio(chemo)therapy for locally advanced or recurrent/metastatic esophageal squamous cell carcinoma. Discov Oncol 2023; 14:165. [PMID: 37665394 PMCID: PMC10477157 DOI: 10.1007/s12672-023-00783-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 08/28/2023] [Indexed: 09/05/2023] Open
Abstract
OBJECTIVE This study was designed to investigate the efficacy and prognostic factors for immune checkpoint inhibitors (ICIs) combined with or without radio(chemo)therapy and to evaluate their toxicity in patients with locally advanced or recurrent/metastatic esophageal squamous cell carcinoma (LA/RM ESCC). METHODS In this study, 198 patients with locally advanced or recurrent/metastatic (LA/RM) ESCC who received ICIs combined with or without radiotherapy/chemotherapy in the Department of Radiotherapy of the Fourth Hospital of Hebei Medical University were retrospectively analyzed. Univariate and multivariate analyses were performed to determine the prognostic factors for overall survival (OS) and progression free survival (PFS). The factors affecting treatment response and the occurrences of treatment-related adverse events (trAEs) were analyzed. RESULTS The median OS and PFS were 30.4 months (95% confidence interval [CI] 15.1-45.7 months) and 15.3 months (95% CI 12.8-17.8 months), respectively. Univariate and multivariate analysis showed that the number of ICI cycles, the intervention of radiotherapy and dysphagia were independent factors affecting OS (Hazard ratio [HR] = 0.39, 2.043 and 0.365, respectively; P = 0.018, 0.001 and 0.032, respectively). The intervention of radiotherapy was an independent factor for PFS (hazard ratio [HR] = 18.149, P = 0.013). The median OS and PFS for patients who had complete response and partial response (Objective response, ORR) were 50.8 months (95% CI 25.8-75.7 months) and 20.5 months (95% CI 14.1-27.0), respectively, which were significantly higher than those in the non-ORR group (OSnon-ORR:17.5 months, 95% CI 14.0-21.0; χ2 = 13.881, P < 0.001; PFSnon-ORR: 12.1 months, 95% CI 10.1-14.1, χ2 = 10.676, P = 0.001). The intervention of radiotherapy could improve treatment response (χ2 = 47.725, P = 0.000). In entire study population, 83 patients (41.9%) had ≥ grade 2 trAEs. CONCLUSIONS ICIs combined with radiotherapy/chemotherapy are safe and effective in LA/RM ESCC patients. Intervention of radiotherapy, the number of immunotherapy cycles and occurrence of dysphagia affecting the overall survival of LR/RM ESCC patients. Intervention of radiotherapy was an independent prognosis factor for OS and PFS and associated with better treatment response.
Collapse
Affiliation(s)
- Xiao-Han Zhao
- Department of Radiation Oncology, The Forth Hospital of Hebei Medical University, No. 12 Jiankan Road, Chang'an District, Shijiazhuang, 050011, China
| | - Hong-Mei Gao
- Department of Radiation, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Jing-Yuan Wen
- Department of Radiation Oncology, The Forth Hospital of Hebei Medical University, No. 12 Jiankan Road, Chang'an District, Shijiazhuang, 050011, China
| | - He-Song Wang
- Department of Radiation Oncology, The Forth Hospital of Hebei Medical University, No. 12 Jiankan Road, Chang'an District, Shijiazhuang, 050011, China
| | - Luan-Ying Wu
- Department of Radiation Oncology, The Forth Hospital of Hebei Medical University, No. 12 Jiankan Road, Chang'an District, Shijiazhuang, 050011, China
| | - Chun-Yang Song
- Department of Radiation Oncology, The Forth Hospital of Hebei Medical University, No. 12 Jiankan Road, Chang'an District, Shijiazhuang, 050011, China
| | - Wen-Zhao Deng
- Department of Radiation Oncology, The Forth Hospital of Hebei Medical University, No. 12 Jiankan Road, Chang'an District, Shijiazhuang, 050011, China
| | - Shu-Chai Zhu
- Department of Radiation Oncology, The Forth Hospital of Hebei Medical University, No. 12 Jiankan Road, Chang'an District, Shijiazhuang, 050011, China
| | - Wen-Bin Shen
- Department of Radiation Oncology, The Forth Hospital of Hebei Medical University, No. 12 Jiankan Road, Chang'an District, Shijiazhuang, 050011, China.
| |
Collapse
|
13
|
Irianto T, Gaipl US, Rückert M. Immune modulation during anti-cancer radio(immuno)therapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 382:239-277. [PMID: 38225105 DOI: 10.1016/bs.ircmb.2023.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
Cancer can affect all human organs and tissues and ranks as a prominent cause of death as well as an obstruction to increasing life expectancy. A notable breakthrough in oncology has been the inclusion of the immune system in fighting cancer, potentially prolonging life and providing long-term benefits. The concept of "immunotherapy" has been discussed from the 19th and early 20th centuries by Wilhelm Busch, William B. Coley and Paul Ehrlich. This involves distinct approaches, including vaccines, non-specific cytokines and adoptive cell therapies. However, despite the advances made in recent years, questions on how to select the best therapeutic options or how to select the best combinations to improve clinical outcomes are still relevant for scientists and clinicians. More than half of cancer patients receive radiotherapy (RT) as part of their treatment. With the advances in RT and immunotherapy approaches, it is reasonable to consider how to enhance immunotherapy with radiation and vice versa, and to investigate whether combinations of these therapies would be beneficial. In this chapter, we will discuss how the immune system responds to cancer cells and different cancer therapies with a focus on combination of RT and immunotherapy (radioimmunotherapy, RIT).
Collapse
Affiliation(s)
- Teresa Irianto
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Udo S Gaipl
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Michael Rückert
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany.
| |
Collapse
|
14
|
Kim Y, Choe BY, Suh TS, Sung W. A Mathematical Model for Predicting Patient Responses to Combined Radiotherapy with CTLA-4 Immune Checkpoint Inhibitors. Cells 2023; 12:cells12091305. [PMID: 37174706 PMCID: PMC10177154 DOI: 10.3390/cells12091305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/27/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
The purpose of this study was to develop a cell-cell interaction model that could predict a tumor's response to radiotherapy (RT) combined with CTLA-4 immune checkpoint inhibition (ICI) in patients with hepatocellular carcinoma (HCC). The previously developed model was extended by adding a new term representing tremelimumab, an inhibitor of CTLA-4. The distribution of the new immune activation term was derived from the results of a clinical trial for tremelimumab monotherapy (NCT01008358). The proposed model successfully reproduced longitudinal tumor diameter changes in HCC patients treated with tremelimumab (complete response = 0%, partial response = 17.6%, stable disease = 58.8%, and progressive disease = 23.6%). For the non-irradiated tumor control group, adding ICI to RT increased the clinical benefit rate from 8% to 32%. The simulation predicts that it is beneficial to start CTLA-4 blockade before RT in terms of treatment sequences. We developed a mathematical model that can predict the response of patients to the combined CTLA-4 blockade with radiation therapy. We anticipate that the developed model will be helpful for designing clinical trials with the ultimate aim of maximizing the efficacy of ICI-RT combination therapy.
Collapse
Affiliation(s)
- Yongjin Kim
- Department of Biomedical Engineering and of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Bo-Young Choe
- Department of Biomedical Engineering and of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Tae Suk Suh
- Department of Biomedical Engineering and of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Wonmo Sung
- Department of Biomedical Engineering and of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|