1
|
Reitmajer M, Nanz L, Müller N, Leiter U, Amaral T, Aebischer V, Flatz L, Forschner A. Comparative real-world outcomes of stage III melanoma patients treated with talimogene laherparepvec or interleukin 2. Ther Adv Med Oncol 2025; 17:17588359251324035. [PMID: 40171522 PMCID: PMC11960150 DOI: 10.1177/17588359251324035] [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: 11/25/2024] [Accepted: 02/12/2025] [Indexed: 04/03/2025] Open
Abstract
Background Talimogene laherparepvec (T-VEC) and interleukin-2 (IL-2) are both used in the intralesional treatment of melanoma skin metastases. T-VEC received regulatory approval from the European Medicines Agency and the U.S. Food and Drug Administration in 2015, while IL-2 has been used off-label for this purpose for many years. Despite their use in clinical practice, there is a lack of comparative data on the efficacy and safety of these treatments. Objectives This retrospective study aimed to compare the efficacy and safety of intralesional T-VEC and IL-2 in non-resectable stage III patients with melanoma treated at a single center between January 2016 and September 2024. Methods We identified eligible patients using the Central Malignant Melanoma Registry and the local University Hospital Pharmacy database. Overall survival (OS) and progression-free survival (PFS) were calculated. Furthermore, best response rates and occurrence of adverse events (AEs) were compared between the T-VEC and the IL-2 group. Concomitant systemic treatment was allowed. Results A total of 62 patients were included, with 37 receiving T-VEC and 25 receiving IL-2 as first-line therapy. Ten patients received both therapies subsequently. The median PFS for the cohort was 5.0 months, and the median OS was 34.0 months. No significant differences in PFS (p = 0.790), OS (p = 0.894), or best response rates (p = 0.468) were found between groups. Common AEs included local injection site reactions and fever, with no severe events leading to discontinuation by a physician. Conclusion No significant differences in PFS, OS, or best response rates were observed between IL-2 and T-VEC treatments. The choice of therapy may be influenced by factors such as availability, physician preference, and patient-specific considerations.
Collapse
Affiliation(s)
- Markus Reitmajer
- Department of Dermatology, University Hospital Tuebingen, Liebermeisterstraße 25, Tuebingen 72076, Germany
| | - Lena Nanz
- Department of Dermatology, University Hospital Tuebingen, Tuebingen, Germany
| | - Nina Müller
- University Pharmacy, University Hospital Tuebingen, Tuebingen, Germany
| | - Ulrike Leiter
- Department of Dermatology, University Hospital Tuebingen, Tuebingen, Germany
| | - Teresa Amaral
- Department of Dermatology, University Hospital Tuebingen, Tuebingen, Germany
| | - Valentin Aebischer
- Department of Dermatology, University Hospital Tuebingen, Tuebingen, Germany
| | - Lukas Flatz
- Department of Dermatology, University Hospital Tuebingen, Tuebingen, Germany
| | - Andrea Forschner
- Department of Dermatology, University Hospital Tuebingen, Tuebingen, Germany
| |
Collapse
|
2
|
Zhang Z, Langenbach M, Sagar S, Fetsch V, Stritzker J, Severa E, Meng K, Winkler F, Rana N, Zoldan K, Godbole I, Solis S, Weber JS, Rafei-Shamsabadi D, Lehr S, Diehl R, Venhoff AC, Voll RE, Buettner N, Neumann-Haefelin C, Boettler T, Hofmann M, Boerries M, Meiss F, Zeiser R, Thimme R, Herati RS, Bengsch B. Efficacy of CTLA-4 checkpoint therapy is dependent on IL-21 signaling to mediate cytotoxic reprogramming of PD-1 +CD8 + T cells. Nat Immunol 2025; 26:92-104. [PMID: 39702858 DOI: 10.1038/s41590-024-02027-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 10/28/2024] [Indexed: 12/21/2024]
Abstract
The mechanisms underlying the efficacy of anti-programmed cell death protein 1 (PD-1) and anti-cytotoxic T lymphocyte-associated protein 4 (CTLA-4) therapy are incompletely understood. Here, by immune profiling responding PD-1+CD8+ T (TResp) cell populations from patients with advanced melanoma, we identified differential programming of TResp cells in response to combination therapy, from an exhausted toward a more cytotoxic effector program. This effect does not occur with anti-PD-1 monotherapy. Single-cell transcriptome and T cell receptor repertoire analysis was used to identify altered effector programming of expanding PD-1+CD8+ T cell clones with distinct regulon usage, STAT1 and STAT3 utilization and antitumor specificity connected to interleukin (IL)-21 signaling in combination and anti-CTLA-4 monotherapy. Therapeutic efficacy of CTLA-4 blockade was lost in B16F10 melanoma models with either Il21r- deficiency or anti-IL-21 receptor blockade. Together, these results show how IL-21 signaling to TResp is critical for anti-CTLA-4-based checkpoint therapies and highlight major signaling differences to anti-PD-1 monotherapy.
Collapse
Affiliation(s)
- Zhen Zhang
- Faculty of Medicine, Clinic for Internal Medicine II, Gastroenterology, Hepatology, Endocrinology and Infectious Disease, University Medical Center Freiburg, Freiburg, Germany
| | - Marlene Langenbach
- Faculty of Medicine, Clinic for Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Sagar Sagar
- Faculty of Medicine, Clinic for Internal Medicine II, Gastroenterology, Hepatology, Endocrinology and Infectious Disease, University Medical Center Freiburg, Freiburg, Germany
| | - Viktor Fetsch
- Faculty of Medicine, Clinic for Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Jonas Stritzker
- Faculty of Medicine, Clinic for Internal Medicine II, Gastroenterology, Hepatology, Endocrinology and Infectious Disease, University Medical Center Freiburg, Freiburg, Germany
| | - Elizabeth Severa
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Ke Meng
- Faculty of Medicine, Clinic for Internal Medicine II, Gastroenterology, Hepatology, Endocrinology and Infectious Disease, University Medical Center Freiburg, Freiburg, Germany
| | - Frances Winkler
- Faculty of Medicine, Clinic for Internal Medicine II, Gastroenterology, Hepatology, Endocrinology and Infectious Disease, University Medical Center Freiburg, Freiburg, Germany
| | - Nisha Rana
- Faculty of Medicine, Clinic for Internal Medicine II, Gastroenterology, Hepatology, Endocrinology and Infectious Disease, University Medical Center Freiburg, Freiburg, Germany
| | - Katharina Zoldan
- Faculty of Medicine, Clinic for Internal Medicine II, Gastroenterology, Hepatology, Endocrinology and Infectious Disease, University Medical Center Freiburg, Freiburg, Germany
| | - Ira Godbole
- Faculty of Medicine, Clinic for Internal Medicine II, Gastroenterology, Hepatology, Endocrinology and Infectious Disease, University Medical Center Freiburg, Freiburg, Germany
| | - Sabrina Solis
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Jeffrey S Weber
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - David Rafei-Shamsabadi
- Department of Dermatology and Venereology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Saskia Lehr
- Department of Dermatology and Venereology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Rebecca Diehl
- Department of Dermatology and Venereology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Ana Cecilia Venhoff
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Reinhard E Voll
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Nico Buettner
- Faculty of Medicine, Clinic for Internal Medicine II, Gastroenterology, Hepatology, Endocrinology and Infectious Disease, University Medical Center Freiburg, Freiburg, Germany
| | - Christoph Neumann-Haefelin
- Faculty of Medicine, Clinic for Internal Medicine II, Gastroenterology, Hepatology, Endocrinology and Infectious Disease, University Medical Center Freiburg, Freiburg, Germany
- Department of Gastroenterology and Hepatology, University Hospital Cologne, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Tobias Boettler
- Faculty of Medicine, Clinic for Internal Medicine II, Gastroenterology, Hepatology, Endocrinology and Infectious Disease, University Medical Center Freiburg, Freiburg, Germany
| | - Maike Hofmann
- Faculty of Medicine, Clinic for Internal Medicine II, Gastroenterology, Hepatology, Endocrinology and Infectious Disease, University Medical Center Freiburg, Freiburg, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Frank Meiss
- Department of Dermatology and Venereology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Faculty of Medicine, Clinic for Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Robert Thimme
- Faculty of Medicine, Clinic for Internal Medicine II, Gastroenterology, Hepatology, Endocrinology and Infectious Disease, University Medical Center Freiburg, Freiburg, Germany
| | - Ramin S Herati
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Bertram Bengsch
- Faculty of Medicine, Clinic for Internal Medicine II, Gastroenterology, Hepatology, Endocrinology and Infectious Disease, University Medical Center Freiburg, Freiburg, Germany.
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.
- German Cancer Consortium (DKTK) Heidelberg, Germany, Partner Site Freiburg, Freiburg, Germany.
| |
Collapse
|
3
|
Stull CM, Clark D, Parker T, Idriss MH, Patel VA, Migden MR. Current and emerging intralesional immunotherapies in cutaneous oncology. J Am Acad Dermatol 2024; 91:910-921. [PMID: 38942173 DOI: 10.1016/j.jaad.2024.05.095] [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: 09/25/2023] [Revised: 05/12/2024] [Accepted: 05/29/2024] [Indexed: 06/30/2024]
Abstract
Immunotherapies have revolutionized the management of advanced cutaneous malignancies. However, some patients fail to respond to these therapies, others are ineligible because of comorbidities, and a minority of patients experience treatment-limiting systemic immune-related adverse events. To address these issues and expand treatment options for patients with early-stage disease, a variety of immunotherapies are being developed for direct intratumoral administration. Agents including oncolytic viruses, monoclonal antibodies, cytokines, peptides, and pattern-recognition receptor agonists have been engineered to evoke a local immune response while minimizing systemic toxicity and have shown favorable results in preclinical and early clinical testing. This review covers the current landscape of intratumoral immunotherapies for the treatment of cutaneous melanoma, squamous cell carcinoma, and basal cell carcinoma, highlighting the diverse array of agents being explored and their potential benefits and challenges.
Collapse
Affiliation(s)
- Carolyn M Stull
- Department of Dermatology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Denise Clark
- Department of Dermatology, University of Cincinnati, Cincinnati, Ohio
| | - Tayler Parker
- Department of Dermatology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Munir H Idriss
- Department of Dermatology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vishal A Patel
- Department of Dermatology, George Washington School of Medicine and Health Sciences, Washington, District of Columbia
| | - Michael R Migden
- Departments of Dermatology and Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
5
|
Lutz EA, Jailkhani N, Momin N, Huang Y, Sheen A, Kang BH, Wittrup KD, Hynes RO. Intratumoral nanobody-IL-2 fusions that bind the tumor extracellular matrix suppress solid tumor growth in mice. PNAS NEXUS 2022; 1:pgac244. [PMID: 36712341 PMCID: PMC9802395 DOI: 10.1093/pnasnexus/pgac244] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022]
Abstract
Confining cytokine exposure to the tumors would greatly enhance cancer immunotherapy safety and efficacy. Immunocytokines, cytokines fused to tumor-targeting antibodies, have been developed with this intention, but without significant clinical success to date. A critical limitation is uptake by receptor-expressing cells in the blood, that decreases the dose at the tumor and engenders toxicity. Small-format immunocytokines, constructed with antibody fragments, are hypothesized to improve tumor specificity due to rapid systemic clearance. However, effective design criteria for small-format immunocytokines need further examination. Here, we engineer small interleukin-2 (IL-2) immunocytokines fused to nanobodies with nanomolar to picomolar affinities for the tumor-specific EIIIB domain of fibronectin (also known as EDB). Upon intravenous delivery into immunocompetent mice, such immunocytokines led to similar tumor growth delay as size-matched untargeted IL-2. Intratumoral (i.t.) delivery imparted improved survival dependent on affinity to EIIIB. I.t. administration offers a promising avenue to deliver small-format immunocytokines, given effective affinity for the tumor microenvironment.
Collapse
Affiliation(s)
| | | | | | - Ying Huang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Allison Sheen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Byong H Kang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | |
Collapse
|