151
|
Liang S, Wang H, Tian H, Xu Z, Wu M, Hua D, Li C. The prognostic biological markers of immunotherapy for non-small cell lung cancer: current landscape and future perspective. Front Immunol 2023; 14:1249980. [PMID: 37753089 PMCID: PMC10518408 DOI: 10.3389/fimmu.2023.1249980] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/24/2023] [Indexed: 09/28/2023] Open
Abstract
The emergence of immunotherapy, particularly programmed cell death 1 (PD-1) and programmed cell death ligand-1 (PD-L1) produced profound transformations for treating non-small cell lung cancer (NSCLC). Nevertheless, not all NSCLC patients can benefit from immunotherapy in clinical practice. In addition to limited response rates, exorbitant treatment costs, and the substantial threats involved with immune-related adverse events, the intricate interplay between long-term survival outcomes and early disease progression, including early immune hyperprogression, remains unclear. Consequently, there is an urgent imperative to identify robust predictive and prognostic biological markers, which not only possess the potential to accurately forecast the therapeutic efficacy of immunotherapy in NSCLC but also facilitate the identification of patient subgroups amenable to personalized treatment approaches. Furthermore, this advancement in patient stratification based on certain biological markers can also provide invaluable support for the management of immunotherapy in NSCLC patients. Hence, in this review, we comprehensively examine the current landscape of individual biological markers, including PD-L1 expression, tumor mutational burden, hematological biological markers, and gene mutations, while also exploring the potential of combined biological markers encompassing radiological and radiomic markers, as well as prediction models that have the potential to better predict responders to immunotherapy in NSCLC with an emphasis on some directions that warrant further investigation which can also deepen the understanding of clinicians and provide a reference for clinical practice.
Collapse
Affiliation(s)
- Shuai Liang
- Department of Oncology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Hanyu Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Haixia Tian
- Department of Oncology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Zhicheng Xu
- Department of Oncology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Min Wu
- Suzhou Cancer Center Core Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Dong Hua
- Department of Oncology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Chengming Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
152
|
Stukalin I, Navani V, Gupta M, Ruan Y, Boyne DJ, O’Sullivan DE, Meyers DE, Goutam S, Sander M, Ewanchuk BW, Brenner DR, Suo A, Cheung WY, Heng DYC, Monzon JG, Cheng T. Development and Validation of a Prognostic Risk Model for Patients with Advanced Melanoma Treated with Immune Checkpoint Inhibitors. Oncologist 2023; 28:812-822. [PMID: 37011230 PMCID: PMC10485285 DOI: 10.1093/oncolo/oyad073] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 02/08/2023] [Indexed: 04/05/2023] Open
Abstract
BACKGROUND Risk stratification tools for patients with advanced melanoma (AM) treated with immune checkpoint inhibitors (ICI) are lacking. We identified a new prognostic model associated with overall survival (OS). PATIENTS AND METHODS A total of 318 treatment naïve patients with AM receiving ICI were collected from a multi-centre retrospective cohort study. LASSO Cox regression identified independent prognostic factors associated with OS. Model validation was carried out on 500 iterations of bootstrapped samples. Harrel's C-index was calculated and internally validated to outline the model's discriminatory performance. External validation was carried out in 142 advanced melanoma patients receiving ICI in later lines. RESULTS High white blood cell count (WBC), high lactate dehydrogenase (LDH), low albumin, Eastern Cooperative Oncology Group (ECOG) performance status ≥1, and the presence of liver metastases were included in the model. Patients were parsed into 3 risk groups: favorable (0-1 factors) OS of 52.9 months, intermediate (2-3 factors) OS 13.0 months, and poor (≥4 factors) OS 2.7 months. The C-index of the model from the discovery cohort was 0.69. External validation in later-lines (N = 142) of therapy demonstrated a c-index of 0.65. CONCLUSIONS Liver metastases, low albumin, high LDH, high WBC, and ECOG≥1 can be combined into a prognostic model for AM patients treated with ICI.
Collapse
Affiliation(s)
- Igor Stukalin
- Department of Oncology, Division of Medical Oncology, Tom Baker Cancer Centre, Calgary, AB, Canada
| | - Vishal Navani
- Department of Oncology, Division of Medical Oncology, Tom Baker Cancer Centre, Calgary, AB, Canada
| | - Mehul Gupta
- Department of Oncology, Division of Medical Oncology, Tom Baker Cancer Centre, Calgary, AB, Canada
| | - Yibing Ruan
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Devon J Boyne
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Dylan E O’Sullivan
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Daniel E Meyers
- Department of Oncology, Division of Medical Oncology, Tom Baker Cancer Centre, Calgary, AB, Canada
| | - Siddhartha Goutam
- Department of Oncology, Division of Medical Oncology, Tom Baker Cancer Centre, Calgary, AB, Canada
| | - Michael Sander
- Department of Oncology, Division of Medical Oncology, Tom Baker Cancer Centre, Calgary, AB, Canada
| | - Benjamin W Ewanchuk
- Department of Oncology, Division of Medical Oncology, Tom Baker Cancer Centre, Calgary, AB, Canada
| | - Darren R Brenner
- Department of Oncology, Division of Medical Oncology, Tom Baker Cancer Centre, Calgary, AB, Canada
| | - Aleksi Suo
- Department of Oncology, Division of Medical Oncology, Tom Baker Cancer Centre, Calgary, AB, Canada
| | - Winson Y Cheung
- Department of Oncology, Division of Medical Oncology, Tom Baker Cancer Centre, Calgary, AB, Canada
| | - Daniel Y C Heng
- Department of Oncology, Division of Medical Oncology, Tom Baker Cancer Centre, Calgary, AB, Canada
| | - Jose G Monzon
- Department of Oncology, Division of Medical Oncology, Tom Baker Cancer Centre, Calgary, AB, Canada
| | - Tina Cheng
- Department of Oncology, Division of Medical Oncology, Tom Baker Cancer Centre, Calgary, AB, Canada
| |
Collapse
|
153
|
Trilla-Fuertes L, Gámez-Pozo A, Prado-Vázquez G, López-Vacas R, Soriano V, Garicano F, Lecumberri MJ, Rodríguez de la Borbolla M, Majem M, Pérez-Ruiz E, González-Cao M, Oramas J, Magdaleno A, Fra J, Martín-Carnicero A, Corral M, Puértolas T, Ramos-Ruiz R, Dittmann A, Nanni P, Fresno Vara JÁ, Espinosa E. Multi-omics Characterization of Response to PD-1 Inhibitors in Advanced Melanoma. Cancers (Basel) 2023; 15:4407. [PMID: 37686682 PMCID: PMC10486782 DOI: 10.3390/cancers15174407] [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: 07/25/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023] Open
Abstract
Immunotherapy improves the survival of patients with advanced melanoma, 40% of whom become long-term responders. However, not all patients respond to immunotherapy. Further knowledge of the processes involved in the response and resistance to immunotherapy is still needed. In this study, clinical paraffin samples from fifty-two advanced melanoma patients treated with anti-PD-1 inhibitors were assessed via high-throughput proteomics and RNA-seq. The obtained proteomics and transcriptomics data were analyzed using multi-omics network analyses based on probabilistic graphical models to identify those biological processes involved in the response to immunotherapy. Additionally, proteins related to overall survival were studied. The activity of the node formed by the proteins involved in protein processing in the endoplasmic reticulum and antigen presentation machinery was higher in responders compared to non-responders; the activity of the immune and inflammatory response node was also higher in those with complete or partial responses. A predictor for overall survival based on two proteins (AMBP and PDSM5) was defined. In summary, the response to anti-PD-1 therapy in advanced melanoma is related to protein processing in the endoplasmic reticulum, and also to genes involved in the immune and inflammatory responses. Finally, a two-protein predictor can define survival in advanced disease. The molecular characterization of the mechanisms involved in the response and resistance to immunotherapy in melanoma leads the way to establishing therapeutic alternatives for patients who will not respond to this treatment.
Collapse
Affiliation(s)
- Lucía Trilla-Fuertes
- Molecular Oncology Laboratory, Hospital Universitario La Paz-IdiPAZ, 28046 Madrid, Spain; (L.T.-F.); (A.G.-P.); (G.P.-V.); (R.L.-V.); (J.Á.F.V.)
| | - Angelo Gámez-Pozo
- Molecular Oncology Laboratory, Hospital Universitario La Paz-IdiPAZ, 28046 Madrid, Spain; (L.T.-F.); (A.G.-P.); (G.P.-V.); (R.L.-V.); (J.Á.F.V.)
- Biomedica Molecular Medicine SL, 28049 Madrid, Spain
| | - Guillermo Prado-Vázquez
- Molecular Oncology Laboratory, Hospital Universitario La Paz-IdiPAZ, 28046 Madrid, Spain; (L.T.-F.); (A.G.-P.); (G.P.-V.); (R.L.-V.); (J.Á.F.V.)
- Biomedica Molecular Medicine SL, 28049 Madrid, Spain
| | - Rocío López-Vacas
- Molecular Oncology Laboratory, Hospital Universitario La Paz-IdiPAZ, 28046 Madrid, Spain; (L.T.-F.); (A.G.-P.); (G.P.-V.); (R.L.-V.); (J.Á.F.V.)
| | - Virtudes Soriano
- Instituto Valenciano de Oncología, 46009 Valencia, Spain;
- Spanish Melanoma Group (GEM), 08024 Barcelona, Spain; (F.G.); (M.J.L.); (M.R.d.l.B.); (M.M.); (E.P.-R.); (M.G.-C.); (J.O.); (A.M.); (J.F.); (M.C.); (T.P.)
| | - Fernando Garicano
- Spanish Melanoma Group (GEM), 08024 Barcelona, Spain; (F.G.); (M.J.L.); (M.R.d.l.B.); (M.M.); (E.P.-R.); (M.G.-C.); (J.O.); (A.M.); (J.F.); (M.C.); (T.P.)
- Hospital de Galdakao, 48960 Galdakao, Spain
| | - M. José Lecumberri
- Spanish Melanoma Group (GEM), 08024 Barcelona, Spain; (F.G.); (M.J.L.); (M.R.d.l.B.); (M.M.); (E.P.-R.); (M.G.-C.); (J.O.); (A.M.); (J.F.); (M.C.); (T.P.)
- Complejo Hospitalario de Navarra, 31008 Pamplona, Spain
| | - María Rodríguez de la Borbolla
- Spanish Melanoma Group (GEM), 08024 Barcelona, Spain; (F.G.); (M.J.L.); (M.R.d.l.B.); (M.M.); (E.P.-R.); (M.G.-C.); (J.O.); (A.M.); (J.F.); (M.C.); (T.P.)
- Hospital de Valme, 41014 Sevilla, Spain
| | - Margarita Majem
- Spanish Melanoma Group (GEM), 08024 Barcelona, Spain; (F.G.); (M.J.L.); (M.R.d.l.B.); (M.M.); (E.P.-R.); (M.G.-C.); (J.O.); (A.M.); (J.F.); (M.C.); (T.P.)
- Hospital de la Santa Creu i Sant Pau, 08001 Barcelona, Spain
| | - Elisabeth Pérez-Ruiz
- Spanish Melanoma Group (GEM), 08024 Barcelona, Spain; (F.G.); (M.J.L.); (M.R.d.l.B.); (M.M.); (E.P.-R.); (M.G.-C.); (J.O.); (A.M.); (J.F.); (M.C.); (T.P.)
- Unidad de Gestión Clínica Intercentros (UGCI) de Oncología Médica, Hospitales Universitarios Regional y Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospitales Universitarios Regional y Virgen de la Victoria, 29010 Málaga, Spain
| | - María González-Cao
- Spanish Melanoma Group (GEM), 08024 Barcelona, Spain; (F.G.); (M.J.L.); (M.R.d.l.B.); (M.M.); (E.P.-R.); (M.G.-C.); (J.O.); (A.M.); (J.F.); (M.C.); (T.P.)
- Hospital Quirón Dexeus, 08028 Barcelona, Spain
| | - Juana Oramas
- Spanish Melanoma Group (GEM), 08024 Barcelona, Spain; (F.G.); (M.J.L.); (M.R.d.l.B.); (M.M.); (E.P.-R.); (M.G.-C.); (J.O.); (A.M.); (J.F.); (M.C.); (T.P.)
- Hospital Universitario de Canarias-San Cristóbal de la Laguna, 38320 Tenerife, Spain
| | - Alejandra Magdaleno
- Spanish Melanoma Group (GEM), 08024 Barcelona, Spain; (F.G.); (M.J.L.); (M.R.d.l.B.); (M.M.); (E.P.-R.); (M.G.-C.); (J.O.); (A.M.); (J.F.); (M.C.); (T.P.)
- Hospital Universitario de Elche y Vega Baja, 03203 Alicante, Spain
| | - Joaquín Fra
- Spanish Melanoma Group (GEM), 08024 Barcelona, Spain; (F.G.); (M.J.L.); (M.R.d.l.B.); (M.M.); (E.P.-R.); (M.G.-C.); (J.O.); (A.M.); (J.F.); (M.C.); (T.P.)
- Hospital Universitario Río Hortega, 47012 Valladolid, Spain
| | - Alfonso Martín-Carnicero
- Spanish Melanoma Group (GEM), 08024 Barcelona, Spain; (F.G.); (M.J.L.); (M.R.d.l.B.); (M.M.); (E.P.-R.); (M.G.-C.); (J.O.); (A.M.); (J.F.); (M.C.); (T.P.)
- Hospital San Pedro, 27347 Logroño, Spain
| | - Mónica Corral
- Spanish Melanoma Group (GEM), 08024 Barcelona, Spain; (F.G.); (M.J.L.); (M.R.d.l.B.); (M.M.); (E.P.-R.); (M.G.-C.); (J.O.); (A.M.); (J.F.); (M.C.); (T.P.)
- Hospital Clínico Lozano Blesa, 50009 Zaragoza, Spain
| | - Teresa Puértolas
- Spanish Melanoma Group (GEM), 08024 Barcelona, Spain; (F.G.); (M.J.L.); (M.R.d.l.B.); (M.M.); (E.P.-R.); (M.G.-C.); (J.O.); (A.M.); (J.F.); (M.C.); (T.P.)
- Hospital Universitario Miguel Servet, 50009 Zaragoza, Spain
| | | | - Antje Dittmann
- Functional Genomics Center Zurich, University/ETH Zurich, 8092 Zurich, Switzerland; (A.D.); (P.N.)
| | - Paolo Nanni
- Functional Genomics Center Zurich, University/ETH Zurich, 8092 Zurich, Switzerland; (A.D.); (P.N.)
| | - Juan Ángel Fresno Vara
- Molecular Oncology Laboratory, Hospital Universitario La Paz-IdiPAZ, 28046 Madrid, Spain; (L.T.-F.); (A.G.-P.); (G.P.-V.); (R.L.-V.); (J.Á.F.V.)
- Biomedica Molecular Medicine SL, 28049 Madrid, Spain
- CIBERONC, ISCIII, 28222 Madrid, Spain
| | - Enrique Espinosa
- Spanish Melanoma Group (GEM), 08024 Barcelona, Spain; (F.G.); (M.J.L.); (M.R.d.l.B.); (M.M.); (E.P.-R.); (M.G.-C.); (J.O.); (A.M.); (J.F.); (M.C.); (T.P.)
- CIBERONC, ISCIII, 28222 Madrid, Spain
- Medical Oncology Service, Hospital Universitario La Paz, 28046 Madrid, Spain
| |
Collapse
|
154
|
Boutros C, Belkadi-Sadou D, Marchand A, Roy S, Routier E, Robert C. Cured or Not? Long-term Outcomes of Immunotherapy Responders. Focus on Melanoma. Curr Oncol Rep 2023; 25:989-996. [PMID: 37266890 DOI: 10.1007/s11912-023-01429-x] [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: 05/03/2023] [Indexed: 06/03/2023]
Abstract
PURPOSE OF REVIEW Immune checkpoint inhibitors (ICIs) demonstrated robust antitumor activity and tolerable safety in advanced melanoma. Data on long-term outcome of patients who benefited from this therapy and who are still free of progression despite ICI discontinuation is now available. We review here the characteristics of long-term ICI responders and address the critical question of cure. RECENT FINDINGS Long-term outcome of patients with metastatic melanoma enrolled in large phase 2 and phase 3 clinical trials evaluating ICI in metastatic melanoma is now available. Durable responses, with more than 6 years of median follow-up, may persist after discontinuation. They occur more frequently in patients who achieved a complete response rather than in patients who had partial response or stable disease. Although long-term clinical benefit is more frequent in patients with high PDL-1 expression and smaller tumor burden, durable response may also be observed regardless of baseline characteristics. In patients with asymptomatic brain metastasis, combined immunotherapy (ipilimumab plus nivolumab) may also lead to long-term remission. Clinical trials confirm the durable antitumor activity of ICI. Although the hope for cure seems reasonable for many patients in this situation, late relapses may occur and no relapse-predictive biomarkers have been identified yet. Long-term responders who relapse can respond to a rechallenge of ICI although data are limited concerning the rate and the duration of this new response.
Collapse
Affiliation(s)
- Céline Boutros
- Dermatology Unit, Department of Medicine, Gustave Roussy Cancer Campus, 114 Rue Edouard Vaillant, 94805, Villejuif, France
- Outpatient Clinic, Department of Medicine, Gustave Roussy Cancer Campus, Villejuif, France
| | - Djaouida Belkadi-Sadou
- Dermatology Unit, Department of Medicine, Gustave Roussy Cancer Campus, 114 Rue Edouard Vaillant, 94805, Villejuif, France
| | - Antoine Marchand
- Dermatology Unit, Department of Medicine, Gustave Roussy Cancer Campus, 114 Rue Edouard Vaillant, 94805, Villejuif, France
| | - Séverine Roy
- Dermatology Unit, Department of Medicine, Gustave Roussy Cancer Campus, 114 Rue Edouard Vaillant, 94805, Villejuif, France
| | - Emilie Routier
- Dermatology Unit, Department of Medicine, Gustave Roussy Cancer Campus, 114 Rue Edouard Vaillant, 94805, Villejuif, France
| | - Caroline Robert
- Dermatology Unit, Department of Medicine, Gustave Roussy Cancer Campus, 114 Rue Edouard Vaillant, 94805, Villejuif, France.
- University Paris-Saclay, Faculty of Medicine, Kremlin-Bicêtre, France.
- INSERM Unit U981, Villejuif, France.
| |
Collapse
|
155
|
Ochenduszko S, García Sanchez J, Fita MJJ, González-Barrallo I, Herrero Colomina J, Mujika K, Beveridge RD, Martínez SR, Lafuente BS, Tomas AC, Jaime AB, Cerezuela Fuentes P, Fra PL, Peeters AG, Meana García JA, García MAA, Altozano JP, Cancela M, Puchades AM, Roca FF, Maiques IM. Characteristics and outcomes of advanced melanoma patients with complete response and elective discontinuation of first-line anti-programmed death-1 monotherapy: A real-world multicentre observational cohort study. Pigment Cell Melanoma Res 2023; 36:388-398. [PMID: 37243929 DOI: 10.1111/pcmr.13093] [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: 11/27/2022] [Revised: 05/07/2023] [Accepted: 05/10/2023] [Indexed: 05/29/2023]
Abstract
Anti-programmed death-1 (anti-PD1) treatment has significantly improved outcomes of advanced melanoma with a considerable percentage of patients achieving complete response (CR). This real-world study analyzed the feasibility of elective anti-PD1 discontinuation in advanced melanoma patients with CR and evaluated factors related to sustained response. Thirty-five patients with advanced cutaneous or primary unknown melanoma with CR to nivolumab or pembrolizumab from 11 centers were included. Mean age was 66.5 years, and 97.1% had ECOG PS 0-1. 28.6% had ≥3 metastatic sites with 58.8% having M1a-M1b disease; 8.6% had liver and 5.7% had brain metastases. At baseline, 80% had normal LDH, and 85.7% had a neutrophil-to-lymphocyte ratio ≤3. 74.3% of patients had CR confirmed in PET-CT. Median duration of anti-PD1 was 23.4 months (range 1.3-50.5). 24 months after therapy discontinuation, 91.9% of patients were progression-free. Estimated PFS and OS at 36, 48, and 60 months from the start of anti-PD1 were 94.2%, 89.9%, 84.3%, and 97.1%, 93.3%, 93.3%, respectively. Antibiotics use after anti-PD1 discontinuation increased the odds of progression (OR 16.53 [95% CI 1.7, 226.03]). The study confirms the feasibility of elective anti-PD1 discontinuation in advanced melanoma patients with CR and favorable prognostic factors at baseline.
Collapse
Affiliation(s)
| | | | | | | | | | - Karmele Mujika
- Unidad del Cáncer de Gipuzkoa, OSID-Onkologikoa, San Sebastian, Spain
| | | | | | | | - Alberto Cunquero Tomas
- Consorcio Hospital General Universitario de Valencia, Hospital General de Requena, Requena, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
156
|
Earland N, Zhang W, Usmani A, Nene A, Bacchiocchi A, Chen DY, Sznol M, Halaban R, Chaudhuri AA, Newman AM. CD4 T cells and toxicity from immune checkpoint blockade. Immunol Rev 2023; 318:96-109. [PMID: 37491734 PMCID: PMC10838135 DOI: 10.1111/imr.13248] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 06/27/2023] [Indexed: 07/27/2023]
Abstract
Immune-related toxicities, otherwise known as immune-related adverse events (irAEs), occur in a substantial fraction of cancer patients treated with immune checkpoint inhibitors (ICIs). Ranging from asymptomatic to life-threatening, ICI-induced irAEs can result in hospital admission, high-dose corticosteroid treatment, ICI discontinuation, and in some cases, death. A deeper understanding of the factors underpinning severe irAE development will be essential for improved irAE prediction and prevention, toward maximizing the benefits and safety profiles of ICIs. In recent work, we applied mass cytometry, single-cell RNA sequencing, single-cell V(D)J sequencing, bulk RNA sequencing, and bulk T-cell receptor (TCR) sequencing to identify pretreatment determinants of severe irAE development in patients with advanced melanoma. Across 71 patients separated into three cohorts, we found that two baseline features in circulation-elevated activated CD4 effector memory T-cell abundance and TCR diversity-are associated with severe irAE development, independent of the affected organ system within 3 months of ICI treatment initiation. Here, we provide an extended perspective on this work, synthesize and discuss related literature, and summarize practical considerations for clinical translation. Collectively, these findings lay a foundation for data-driven and mechanistic insights into irAE development, with the potential to reduce ICI morbidity and mortality in the future.
Collapse
Affiliation(s)
- Noah Earland
- Division of Cancer Biology, Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Wubing Zhang
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Abul Usmani
- Division of Cancer Biology, Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Antonella Bacchiocchi
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
| | - David Y. Chen
- Division of Dermatology, Washington University School of Medicine, St. Louis, MO, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Mario Sznol
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Medicine, Division of Medical Oncology, Yale University School of Medicine, New Haven, CT, USA
| | - Ruth Halaban
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
| | - Aadel A. Chaudhuri
- Division of Cancer Biology, Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
- Department of Computer Science and Engineering, Washington University in St. Louis, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Aaron M. Newman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| |
Collapse
|
157
|
Lee S, Bennett AV, Zhou X, Betof Warner A, Trogdon JG, Kent EE, Lund JL. Real-world treatment patterns and outcomes for patients with advanced melanoma treated with immunotherapy or targeted therapy. Pharmacoepidemiol Drug Saf 2023; 32:988-1000. [PMID: 37095605 DOI: 10.1002/pds.5630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 03/17/2023] [Accepted: 04/12/2023] [Indexed: 04/26/2023]
Abstract
OBJECTIVE To identify real-world patterns of first line treatment, treatment sequence and outcomes for older adults diagnosed with advanced melanoma who received immunotherapy or targeted therapy. METHODS The study population included older adults (ages 65+) diagnosed with unresectable or metastatic melanoma between 2012 and 2017 and who received first line immunotherapy or targeted therapy. Using the linked surveillance, epidemiology, and end results-medicare data, we described patterns of first line treatment and treatment sequence through 2018. We used descriptive statistics to report patient and provider characteristics by first line treatment receipt and changes in first line therapy use over calendar time. We also described overall survival (OS) and time to treatment failure (TTF) by first line treatment using the Kaplan-Meier method. For patterns of treatment sequence, we reported commonly observed treatment switch patterns by treatment sub-category and calendar year. RESULTS The analyses included 584 patients (mean age = 76.3 years). A majority (n = 502) received first line immunotherapy. There was a sustained increase in immunotherapy uptake, most notably from 2015 to 2016. The estimated median OS and TTF were longer with first line immunotherapy than with targeted therapy. Individuals treated with CTLA-4 + PD-1 inhibitors had the longest median OS (28.4 months). The most common treatment switch pattern was from a first line CTLA-4 inhibitor to a second line PD-1 inhibitor. CONCLUSIONS Our findings inform understanding of treatment patterns of currently used immunotherapies and targeted therapies in older adults with advanced melanoma. Immunotherapy use has increased steadily with PD-1 inhibitors becoming a dominant treatment option since 2015.
Collapse
Affiliation(s)
- Sejin Lee
- Department of Health Policy and Management, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Antonia V Bennett
- Department of Health Policy and Management, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Xi Zhou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - Justin G Trogdon
- Department of Health Policy and Management, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Erin E Kent
- Department of Health Policy and Management, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jennifer L Lund
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
158
|
Sharma H, Moturi KR, Pankratz VS, Yilmaz E, Gbolahan OB, Kumar A, Hashemi-Sadraei N. Outcomes of responders to PD-1/PD-L1 inhibitors who discontinue therapy after sustained disease control. J Cancer Res Clin Oncol 2023; 149:8673-8680. [PMID: 37115270 PMCID: PMC10140701 DOI: 10.1007/s00432-023-04812-0] [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: 02/02/2023] [Accepted: 04/22/2023] [Indexed: 04/29/2023]
Abstract
BACKGROUND PD-1/PD-L1 immune checkpoint inhibitors (ICIs) are widely used in the treatment of metastatic malignancies. Judiciously balancing disease control (DC) against development of immune-related adverse events (irAE) remains a crucial aspect of treatment. The effect of treatment discontinuation after sustained disease control (SDC) is unknown. The purpose of this analysis was to evaluate outcomes of responders to ICI who discontinue treatment after a minimum of 12 months (SDC). METHODS We retrospectively reviewed the database of the University of New Mexico Comprehensive Cancer Center (UNMCCC) between 2014 and 2021 and identified patients who had received ICI. Patients with metastatic solid tumors who had stopped ICI therapy after achieving SDC [stable disease, partial response, complete response (SD, PR, CR)] were selected and outcomes reviewed from their electronic health records. RESULTS We identified 204 patients who were treated with ICI for various solid cancers. Forty-four patients (21.6%) met the criteria, of whom 35 with follow-up data were included in the final analysis; including 11 melanoma, 5 non-small cell lung, 4 head & neck, 8 renal, 4 urothelial, 1 anal, 1 Merkel cell carcinoma, and 1 liposarcoma. Patients were divided into two groups: those who stopped ICI due to an irAE [irAE group, n = 14, median treatment time (MTT), 16.6 mo] and those who stopped due to other reasons (eg completion of 2 years of therapy, n = 20, non-cancer related surgery, n = 1) (non-irAE group, n = 21, MTT, 23.7 mo). Among the irAE group, the most common irAE included pneumonitis, rash, transaminitis, and fatigue. As of data cutoff date, 9 of 14 (64%) patients continued to show SDC. Only 5 of 14 (36%) patients in this group experienced progression of disease (PD), with 1 of 2 patients achieving DC (median follow-up of 19.2 mo after last dose of treatment, range 3-50.2 mo). Among the non-irAE group, 13 of 21 (62%) continued to have SDC. Eight of 21 (38%) experienced PD after stopping treatment, 7 of whom received ICI rechallenge, with 2 of 7 achieving DC (median follow-up of 22.2 mo, range 3.6-54.8 mo). At a median follow-up of 21.3 mo from stopping ICI therapy (range, 3-54.8 mo), 10 patients (71%) from the irAE group and 13 (61.9%) from the non-irAE group are in DC and have not experienced PD. CONCLUSIONS We demonstrate that 22 (66%) patients experienced SDC, regardless of cancer type or development of irAE. After including patients who were re-challenged with ICI due to PD, 25 (71%) remain in DC. Future prospective malignancy-specific trials are warranted to evaluate optimal treatment duration.
Collapse
Affiliation(s)
- Harsh Sharma
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131 USA
| | - Krishna R. Moturi
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131 USA
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87102 USA
| | - Vernon S. Pankratz
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131 USA
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87102 USA
| | - Emrullah Yilmaz
- Cleveland Clinic Taussig Cancer Center, Cleveland, OH 44106 USA
| | - Olumide B. Gbolahan
- Winship Cancer Institute at Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Atul Kumar
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131 USA
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87102 USA
| | - Neda Hashemi-Sadraei
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131 USA
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87102 USA
| |
Collapse
|
159
|
Schuster C, Akslen LA, Straume O. β2-adrenergic receptor expression in patients receiving bevacizumab therapy for metastatic melanoma. Cancer Med 2023; 12:17891-17900. [PMID: 37551424 PMCID: PMC10524038 DOI: 10.1002/cam4.6424] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 07/01/2023] [Accepted: 07/27/2023] [Indexed: 08/09/2023] Open
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF) was initially known as vascular permeability factor and identified as a driver of tumour angiogenesis. Recently, its role in supporting an immunosuppressive tumour microenvironment was demonstrated, and anti-VEGF treatment combined with immune checkpoint blockade is currently investigated. Further, beta-adrenergic signalling as a modifier of cancer hallmarks like immune response, angiogenesis and metastasis gained increased attention during past years. METHODS Focusing on the aspect of immunosuppression in upregulated beta-adrenergic signalling, we investigated predictive markers in patients with metastatic melanoma who received bevacizumab monotherapy, a specific VEGF-A binding antibody. We explored the expression of beta-2 adrenergic receptor (β2-AR), interleukin 6-receptor (IL6-R), cyclooxygenase 2 (COX2) and VEGF-A by immunohistochemistry in melanoma to assess the correlation between these proteins in melanoma cells and response to treatment. RESULTS Strong β2-AR expression in metastases was associated with clinical benefit of bevacizumab. Furthermore, expression of the latter was positively linked to expression of VEGF-A and COX2. β2-AR expression in melanoma metastasis appears to distinguish a subgroup of patients that might benefit from anti-VEGF treatment. CONCLUSION Our results strengthen further exploration of anti-VEGF therapy in combination with immune checkpoint blockade in clinical studies and the investigation of β2-AR as predictive marker.
Collapse
Affiliation(s)
- Cornelia Schuster
- Department of Clinical Science, Centre for Cancer Biomarkers CCBIOUniversity of BergenBergenNorway
- Department of Oncology and Medical PhysicsHaukeland University HospitalBergenNorway
| | - Lars A. Akslen
- Department of Clinical Medicine, Centre for Cancer Biomarkers CCBIOUniversity of BergenBergenNorway
- Department of PathologyHaukeland University HospitalBergenNorway
| | - Oddbjørn Straume
- Department of Clinical Science, Centre for Cancer Biomarkers CCBIOUniversity of BergenBergenNorway
- Department of Oncology and Medical PhysicsHaukeland University HospitalBergenNorway
| |
Collapse
|
160
|
Yiong CS, Lin TP, Lim VY, Toh TB, Yang VS. Biomarkers for immune checkpoint inhibition in sarcomas - are we close to clinical implementation? Biomark Res 2023; 11:75. [PMID: 37612756 PMCID: PMC10463641 DOI: 10.1186/s40364-023-00513-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 07/26/2023] [Indexed: 08/25/2023] Open
Abstract
Sarcomas are a group of diverse and complex cancers of mesenchymal origin that remains poorly understood. Recent developments in cancer immunotherapy have demonstrated a potential for better outcomes with immune checkpoint inhibition in some sarcomas compared to conventional chemotherapy. Immune checkpoint inhibitors (ICIs) are key agents in cancer immunotherapy, demonstrating improved outcomes in many tumor types. However, most patients with sarcoma do not benefit from treatment, highlighting the need for identification and development of predictive biomarkers for response to ICIs. In this review, we first discuss United States (US) Food and Drug Administration (FDA)-approved and European Medicines Agency (EMA)-approved biomarkers, as well as the limitations of their use in sarcomas. We then review eight potential predictive biomarkers and rationalize their utility in sarcomas. These include gene expression signatures (GES), circulating neutrophil-to-lymphocyte ratio (NLR), indoleamine 2,3-dioxygenase (IDO), lymphocyte activation gene 3 (LAG-3), T cell immunoglobin and mucin domain-containing protein 3 (TIM-3), TP53 mutation status, B cells, and tertiary lymphoid structures (TLS). Finally, we discuss the potential for TLS as both a predictive and prognostic biomarker for ICI response in sarcomas to be implemented in the clinic.
Collapse
Affiliation(s)
- Chin Sern Yiong
- Translational Precision Oncology Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore
- Department of Pharmacy, National University of Singapore, Singapore, 117544, Singapore
| | - Tzu Ping Lin
- Translational Precision Oncology Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore
- Department of Pharmacy, National University of Singapore, Singapore, 117544, Singapore
| | - Vivian Yujing Lim
- Translational Precision Oncology Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore
| | - Tan Boon Toh
- The N.1 Institute for Health, National University of Singapore, Singapore, Singapore
- The Institute for Digital Medicine (WisDM), National University of Singapore, Singapore, Singapore
| | - Valerie Shiwen Yang
- Translational Precision Oncology Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore.
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, 169610, Singapore.
- Duke-NUS Medical School, Oncology Academic Clinical Program, Singapore, 169857, Singapore.
| |
Collapse
|
161
|
Li L, Tianrui K, Chunlei L, Zhendong Q, Xiaoyan C, Wenhong D. HYDIN mutation status as a potential predictor of immune checkpoint inhibitor efficacy in melanoma. Aging (Albany NY) 2023; 15:7997-8012. [PMID: 37595251 PMCID: PMC10496993 DOI: 10.18632/aging.204925] [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: 05/08/2023] [Accepted: 07/10/2023] [Indexed: 08/20/2023]
Abstract
BACKGROUND The advent of immune checkpoint inhibitors (ICIs) has altered the outlook for cancer treatment. The estimation of predictive biomarkers could contribute to maximizing the benefits from ICIs treatment. Here, we explored the association between HYDIN mutations (HYDIN-MUT) in melanoma and ICIs efficacy. METHODS Clinical data and sequencing data from published studies were utilized to assess the association between HYDIN-MUT and the efficacy of ICIs treatment in melanoma patients. RESULTS Compared to other tumor types, HYDIN (36.14%) has the highest mutation rate in melanoma patients. In the anti-PD-1 treated cohort (n = 254), the HYDIN-MUT patients had a longer OS after ICIs treatment than the HYDIN wild-type (HYDIN-WT) patients (HR = 0.590 [95% CI, 0.410-0.847], P = 0.004); the objective response rate (ORR) and durable clinical benefit (DCB) were increased in patients with HYDIN-MUT (ORR = 46.25, DCB = 56.00%) compared to patients with HYDIN-WT (ORR = 30.99%, DCB = 42.76%) (ORR: P = 0.019; DCB: P = 0.060). In the anti-CTLA4 treated cohort (n = 174), HYDIN-MUT patients achieved significantly longer OS than HYDIN-WT patients (HR = 0.549 [95% CI, 0.366-0.823], P = 0.003); the proportion of ORR and DCB in HYDIN-MUT patients was significantly higher than that in HYDIN-WT patients (ORR 40.54% vs. 14.42%, P = 0.031; DCB 45.76% vs. 22.22%, P = 0.002). Further gene set enrichment analysis demonstrated that DNA repair and anti-tumor immunity were significantly enhanced in HYDIN-MUT patients. CONCLUSIONS HYDIN mutations are a potential predictive biomarker of ICIs efficacy in melanoma patients.
Collapse
Affiliation(s)
- Liu Li
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Kuang Tianrui
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Li Chunlei
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Qiu Zhendong
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Chen Xiaoyan
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Deng Wenhong
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| |
Collapse
|
162
|
Dirks I, Keyaerts M, Dirven I, Neyns B, Vandemeulebroucke J. Development and Validation of a Predictive Model for Metastatic Melanoma Patients Treated with Pembrolizumab Based on Automated Analysis of Whole-Body [ 18F]FDG PET/CT Imaging and Clinical Features. Cancers (Basel) 2023; 15:4083. [PMID: 37627111 PMCID: PMC10452475 DOI: 10.3390/cancers15164083] [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: 07/04/2023] [Revised: 08/04/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Antibodies that inhibit the programmed cell death protein 1 (PD-1) receptor offer a significant survival benefit, potentially cure (i.e., durable disease-free survival following treatment discontinuation), a substantial proportion of patients with advanced melanoma. Most patients however fail to respond to such treatment or acquire resistance. Previously, we reported that baseline total metabolic tumour volume (TMTV) determined by whole-body [18F]FDG PET/CT was independently correlated with survival and able to predict the futility of treatment. Manual delineation of [18F]FDG-avid lesions is however labour intensive and not suitable for routine use. A predictive survival model is proposed based on automated analysis of baseline, whole-body [18F]FDG images. METHODS Lesions were segmented on [18F]FDG PET/CT using a deep-learning approach and derived features were investigated through Kaplan-Meier survival estimates with univariate logrank test and Cox regression analyses. Selected parameters were evaluated in multivariate Cox survival regressors. RESULTS In the development set of 69 patients, overall survival prediction based on TMTV, lactate dehydrogenase levels and presence of brain metastases achieved an area under the curve of 0.78 at one year, 0.70 at two years. No statistically significant difference was observed with respect to using manually segmented lesions. Internal validation on 31 patients yielded scores of 0.76 for one year and 0.74 for two years. CONCLUSIONS Automatically extracted TMTV based on whole-body [18F]FDG PET/CT can aid in building predictive models that can support therapeutic decisions in patients treated with immune-checkpoint blockade.
Collapse
Affiliation(s)
- Ine Dirks
- Department of Electronics and Informatics (ETRO), Vrije Universiteit Brussel (VUB), 1050 Brussels, Belgium;
- IMEC, 3001 Leuven, Belgium
| | - Marleen Keyaerts
- Department of Nuclear Medicine, Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel (VUB), 1050 Brussels, Belgium;
| | - Iris Dirven
- Department of Medical Oncology, Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel (VUB), 1050 Brussels, Belgium; (I.D.); (B.N.)
| | - Bart Neyns
- Department of Medical Oncology, Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel (VUB), 1050 Brussels, Belgium; (I.D.); (B.N.)
| | - Jef Vandemeulebroucke
- Department of Electronics and Informatics (ETRO), Vrije Universiteit Brussel (VUB), 1050 Brussels, Belgium;
- IMEC, 3001 Leuven, Belgium
- Department of Radiology, Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel (VUB), 1050 Brussels, Belgium
| |
Collapse
|
163
|
Zhou X, Xu R, Lu T, Xu R, Wang C, Peng B, Chang X, Shen Z, Wang K, Shi J, Zhao J, Zhang LY. Identification of immunotherapy biomarkers for improving the clinical outcome of homologous recombination deficiency patients with lung adenocarcinoma. Aging (Albany NY) 2023; 15:8090-8112. [PMID: 37578930 PMCID: PMC10496994 DOI: 10.18632/aging.204957] [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: 05/17/2023] [Accepted: 07/20/2023] [Indexed: 08/16/2023]
Abstract
Homologous recombination deficiency (HRD) is a common molecular signature of genomic instability and has been shown to be a biomarker for targeted therapies. However, there is a lack of studies on the role of HRD changes in lung adenocarcinoma (LUAD) transcriptomics. HRD scores were determined using single nucleotide polymorphism (SNP) array data from LUAD patients from The Cancer Genome Atlas (TCGA) database. Transcriptional data from patients with different scores were analyzed to identify biomarkers associated with HRD. Candidate biomarkers were validated using Gene Expression Omnibus (GEO)-sourced datasets and an immunotherapy cohort. According to the bulk transcriptome and clinical characteristics of 912 LUAD patients and Single-cell RNA-seq of 9 LUAD patients from TCGA and GEO databases, we observed increased MS4A6A expression in HRD tumors; high MS4A6A expression predicted improved survival outcomes. Furthermore, a comprehensive analysis of the tumor immune microenvironment (TIME) revealed a positive correlation between MS4A6A expression and neoantigen loading and immune cell infiltration. Additionally, the immunotherapy cohort confirmed the possibility of using MS4A6A as a biomarker. Collectively, we suggest that MS4A6A is associated with HRD and provide a new perspective toward identifying promising biomarkers for immunotherapy.
Collapse
Affiliation(s)
- Xiang Zhou
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150084, China
| | - Rongjian Xu
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266005, China
| | - Tong Lu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150084, China
| | - Ran Xu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150084, China
| | - Chenghao Wang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150084, China
| | - Bo Peng
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150084, China
| | - Xiaoyan Chang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150084, China
| | - Zhiping Shen
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150084, China
| | - Kaiyu Wang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150084, China
| | - Jiaxin Shi
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150084, China
| | - Jiaying Zhao
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150084, China
| | - Lin-You Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150084, China
| |
Collapse
|
164
|
Mori T, Namikawa K, Yamazaki N, Kiniwa Y, Yamasaki O, Yoshikawa S, Inozume T, Kato H, Nakai Y, Fukushima S, Takenouchi T, Maekawa T, Matsushita S, Otsuka A, Nomura M, Baba N, Isei T, Saito S, Fujimoto N, Tanaka R, Kaneko T, Kuwatsuka Y, Matsuya T, Nagase K, Onishi M, Onuma T, Nakamura Y. Efficacy of salvage therapies for advanced acral melanoma after anti-PD-1 monotherapy failure: a multicenter retrospective study of 108 Japanese patients. Front Med (Lausanne) 2023; 10:1229937. [PMID: 37636577 PMCID: PMC10448186 DOI: 10.3389/fmed.2023.1229937] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/31/2023] [Indexed: 08/29/2023] Open
Abstract
Background Anti-programmed cell death protein 1 (PD-1) monotherapy is one of the standard systemic therapies for advanced melanoma; however, the efficacy of salvage systemic therapies after PD-1 monotherapy failure (PD-1 MF), particularly in acral melanoma (AM), the main clinical melanoma type in Japanese patients, is unclear. This study aimed to investigate the efficacy of salvage systemic therapies in Japanese patients with AM after PD-1 MF. Patients and methods The study included 108 patients with advanced AM (palm and sole, 72; nail apparatus, 36) who underwent salvage systemic therapy at 24 Japanese institutions. We mainly assessed the objective response rate (ORR), progression-free survival (PFS), and overall survival (OS). Results Thirty-six (33%) patients received ipilimumab, 23 (21%) received nivolumab and ipilimumab (nivo/ipi), 10 (9%) received cytotoxic chemotherapy, 4 (4%) received BRAF and MEK inhibitors (BRAFi/MEKi), and the remaining 35 (32%) continued with PD-1 monotherapy after disease progression. The ORRs in the ipilimumab, nivo/ipi, cytotoxic chemotherapy, and BRAFi/MEKi groups were 8, 17, 0, and 100%, respectively. The nivo/ipi group showed the longest OS (median, 18.9 months); however, differences in ORR, PFS, and OS between the groups were insignificant. The OS in the nivo/ipi group was higher in the palm and sole groups than in the nail apparatus group (median: not reached vs. 8.7 months, p < 0.001). Cox multivariate analysis demonstrated that nail apparatus melanoma independently predicted unfavorable PFS and OS (p = 0.006 and 0.001). The total OS (from PD-1 monotherapy initiation to death/last follow-up) was insignificant between the groups. Conclusion Nivo/ipi was not more effective than cytotoxic chemotherapy and ipilimumab after PD-1 MF in patients with advanced AM. The prognosis after PD-1 MF would be poorer for nail apparatus melanoma than for palm and sole melanoma.
Collapse
Affiliation(s)
- Tatsuhiko Mori
- Department of Skin Oncology/Dermatology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Kenjiro Namikawa
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Naoya Yamazaki
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yukiko Kiniwa
- Department of Dermatology, Shinshu University, Matsumoto, Japan
| | - Osamu Yamasaki
- Department of Dermatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | | | | | - Hiroshi Kato
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yasuo Nakai
- Department of Dermatology, Mie University, Mie, Japan
| | - Satoshi Fukushima
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Tatsuya Takenouchi
- Department of Dermatology, Niigata Cancer Center Hospital, Niigata, Japan
| | - Takeo Maekawa
- Department of Dermatology, Jichi Medical University, Tochigi, Japan
| | - Shigeto Matsushita
- Department of Dermato-Oncology/Dermatology, National Hospital Organization Kagoshima Medical Center, Kagoshima, Japan
| | - Atsushi Otsuka
- Department of Dermatology, Kyoto University, Kyoto, Japan
- Department of Dermatology, Kindai University Hospital, Osaka, Japan
| | - Motoo Nomura
- Department of Clinical Oncology, Kyoto University, Kyoto, Japan
| | - Natsuki Baba
- Department of Dermatology, University of Fukui, Fukui, Japan
| | - Taiki Isei
- Department of Dermatologic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Shintaro Saito
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Noriki Fujimoto
- Department of Dermatology, Shiga University of Medical Science, Otsu, Japan
| | - Ryo Tanaka
- Department of Dermatology, Kawasaki Medical School, Kurashiki, Japan
| | - Takahide Kaneko
- Department of Dermatology, Juntendo University Urayasu Hospital, Chiba, Japan
| | | | - Taisuke Matsuya
- Department of Dermatology, Asahikawa Medical University, Asahikawa, Japan
| | - Kotaro Nagase
- Division of Dermatology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Masazumi Onishi
- Department of Dermatology, Iwate Medical University, Iwate, Japan
| | - Takehiro Onuma
- Department of Dermatology, University of Yamanashi, Yamanashi, Japan
| | - Yasuhiro Nakamura
- Department of Skin Oncology/Dermatology, Saitama Medical University International Medical Center, Saitama, Japan
| |
Collapse
|
165
|
Eikenes G, Liszkay G, Balatoni T, Czirbesz K, Hunyadi K, Kozéki Z, Kispál MT, Baranyai F, Danyi T, Bőcs K, Kenessey I. Therapeutic and Adverse Effect of Anti-PD1 Immunotherapy in Melanoma: A Retrospective, Single-Institute Study of 222 Patients. Cancers (Basel) 2023; 15:3966. [PMID: 37568785 PMCID: PMC10417734 DOI: 10.3390/cancers15153966] [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: 06/30/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND The introduction of immuno- and targeted therapeutic modalities meant a breakthrough step in the therapy of melanoma. As a checkpoint inhibitor, the more effective and less toxic anti-PD1 therapy followed an anti-CTLA4 approach. METHODS From our patient pool, 222 advanced melanoma cases were selected, where anti-PD1 (pembrolizumab, nivolumab) therapy was initiated between March 2015 and December 2020. During our retrospective analysis, the efficacy and safety of the therapy were assessed. RESULTS The median follow-up was 16 months (interval: 0-64 months), and 150 patients (67.6%) received therapy in the first line, while second and third line therapy was performed among 72 patients (32.4%) for the median of 7.0 months (0-60). In 50 cases, BRAF mutations were detected. Ninety-six patients showed objective response (11.3% CR, 32.0% PR). The median PFS was 10.0 months (0-60), and the median OS was 23.0 months (0-64). Autoimmune side effects were found in 79 patients (35.5%); grade 3 occurred in 6.3% of the cases, while 1 patient died due to fulminant pneumonitis (0.25%). CONCLUSION Although the range of immunotherapeutic options is getting wider, in the management of melanoma patients, anti-PD1 monotherapy remains an important, effective, and safe method. However, significant correlation was found between the immune-related side effects and therapeutic efficacy.
Collapse
Affiliation(s)
- Grethe Eikenes
- Department of Dermato-Oncology, National Institute of Oncology, H1122 Budapest, Hungary; (G.E.); (T.B.); (K.C.); (K.H.); (Z.K.); (M.T.K.); (F.B.); (T.D.)
| | - Gabriella Liszkay
- Department of Dermato-Oncology, National Institute of Oncology, H1122 Budapest, Hungary; (G.E.); (T.B.); (K.C.); (K.H.); (Z.K.); (M.T.K.); (F.B.); (T.D.)
- National Tumor Laboratory Project, H1122 Budapest, Hungary;
| | - Tímea Balatoni
- Department of Dermato-Oncology, National Institute of Oncology, H1122 Budapest, Hungary; (G.E.); (T.B.); (K.C.); (K.H.); (Z.K.); (M.T.K.); (F.B.); (T.D.)
| | - Kata Czirbesz
- Department of Dermato-Oncology, National Institute of Oncology, H1122 Budapest, Hungary; (G.E.); (T.B.); (K.C.); (K.H.); (Z.K.); (M.T.K.); (F.B.); (T.D.)
- National Tumor Laboratory Project, H1122 Budapest, Hungary;
| | - Karen Hunyadi
- Department of Dermato-Oncology, National Institute of Oncology, H1122 Budapest, Hungary; (G.E.); (T.B.); (K.C.); (K.H.); (Z.K.); (M.T.K.); (F.B.); (T.D.)
| | - Zsófia Kozéki
- Department of Dermato-Oncology, National Institute of Oncology, H1122 Budapest, Hungary; (G.E.); (T.B.); (K.C.); (K.H.); (Z.K.); (M.T.K.); (F.B.); (T.D.)
| | - Mihály Tamás Kispál
- Department of Dermato-Oncology, National Institute of Oncology, H1122 Budapest, Hungary; (G.E.); (T.B.); (K.C.); (K.H.); (Z.K.); (M.T.K.); (F.B.); (T.D.)
- National Tumor Laboratory Project, H1122 Budapest, Hungary;
| | - Fanni Baranyai
- Department of Dermato-Oncology, National Institute of Oncology, H1122 Budapest, Hungary; (G.E.); (T.B.); (K.C.); (K.H.); (Z.K.); (M.T.K.); (F.B.); (T.D.)
| | - Tímea Danyi
- Department of Dermato-Oncology, National Institute of Oncology, H1122 Budapest, Hungary; (G.E.); (T.B.); (K.C.); (K.H.); (Z.K.); (M.T.K.); (F.B.); (T.D.)
| | - Katalin Bőcs
- Department of Diagnostic Radiology, National Institute of Oncology, H1122 Budapest, Hungary;
| | - István Kenessey
- National Tumor Laboratory Project, H1122 Budapest, Hungary;
- National Cancer Registry, National Institute of Oncology, H1122 Budapest, Hungary
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, H1091 Budapest, Hungary
| |
Collapse
|
166
|
Rizzetto G, De Simoni E, Molinelli E, Offidani A, Simonetti O. Efficacy of Pembrolizumab in Advanced Melanoma: A Narrative Review. Int J Mol Sci 2023; 24:12383. [PMID: 37569757 PMCID: PMC10419154 DOI: 10.3390/ijms241512383] [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: 06/22/2023] [Revised: 07/29/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Pembrolizumab has been shown to increase survival in patients with metastatic melanoma. Considering the numerous oncoming studies, we decided to conduct a narrative review of the latest efficacy evidence regarding the use of pembrolizumab, alone or in combination, in patients with metastatic melanoma. A search was conducted in PubMed using "pembrolizumab," and "metastatic melanoma" as keywords, considering studies from 2022 onward. We reviewed pembrolizumab and associations, cost-effectiveness, virus, advanced acral melanoma, long-term outcomes, real-life data, biomarkers, obesity, and vaccines. In conclusion, pembrolizumab is a fundamental option in the therapy of metastatic melanoma. However, a certain group of patients do not respond and, therefore, new combination options need to be evaluated. In particular, the use of vaccines tailored to tumor epitopes could represent a breakthrough in the treatment of resistant forms. Further studies with larger sample numbers are needed to confirm the preliminary results.
Collapse
Affiliation(s)
| | | | | | | | - Oriana Simonetti
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy; (G.R.); (E.D.S.); (E.M.); (A.O.)
| |
Collapse
|
167
|
Dirven I, Vander Mijnsbrugge AS, Mignon S, Tijtgat J, Kint N, Neyns B. Auto-immune hemolytic anemia and hemophagocytic lymphohistiocytosis as immune-related adverse event in patients with metastatic melanoma and concurrent chronic lymphocytic leukemia: a case series and literature review. Melanoma Res 2023; 33:338-344. [PMID: 37114670 DOI: 10.1097/cmr.0000000000000895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Auto-immune hemolytic anemia (AIHA) and hemophagocytic lymphohistiocytosis (HLH) are both rare immune-related adverse events (irAEs) following treatment with immune checkpoint inhibitors. Consensus treatment guidelines are currently lacking. Patients with a solid malignancy and a concurrent lymphoproliferative disorder, such as chronic lymphocytic leukemia (CLL), might be more prone to develop hematological irAEs. We report the case history of two patients, diagnosed with CLL, who during treatment for metastatic melanoma with nivolumab, a PD-1 immune checkpoint blocking mAb, developed AIHA and HLH in combination with AIHA. Furthermore, we provide a review of the literature on published cases of immune-related AIHA and HLH and their correlation with CLL.
Collapse
MESH Headings
- Humans
- Melanoma/complications
- Leukemia, Lymphocytic, Chronic, B-Cell/complications
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Lymphohistiocytosis, Hemophagocytic/chemically induced
- Lymphohistiocytosis, Hemophagocytic/complications
- Skin Neoplasms
- Anemia, Hemolytic
- Neoplasms, Second Primary
- Anemia, Hemolytic, Autoimmune/chemically induced
Collapse
Affiliation(s)
- Iris Dirven
- Department of Medical Oncology, Vrije Universiteit Brussel (VUB)/Universitair Ziekenhuis Brussel (UZ Brussel)
| | - An-Sofie Vander Mijnsbrugge
- Department of Medical Oncology, Vrije Universiteit Brussel (VUB)/Universitair Ziekenhuis Brussel (UZ Brussel)
| | - Sacha Mignon
- Department of Medical Oncology, Vrije Universiteit Brussel (VUB)/Universitair Ziekenhuis Brussel (UZ Brussel)
| | - Jens Tijtgat
- Department of Medical Oncology, Vrije Universiteit Brussel (VUB)/Universitair Ziekenhuis Brussel (UZ Brussel)
| | - Nicolas Kint
- Department of Hematology, Vrije Universiteit Brussel (VUB)/Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Bart Neyns
- Department of Medical Oncology, Vrije Universiteit Brussel (VUB)/Universitair Ziekenhuis Brussel (UZ Brussel)
| |
Collapse
|
168
|
Lo J, Hanania HL, Keiser MF, Patel AB. Immune checkpoint inhibitor-induced vitiligo in cancer patients: characterization and management. Arch Dermatol Res 2023; 315:1697-1703. [PMID: 36809408 DOI: 10.1007/s00403-023-02577-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 01/04/2023] [Accepted: 02/08/2023] [Indexed: 02/23/2023]
Abstract
This study highlights the range of non-melanoma cancers where ICI-induced vitiligo can be present and challenges the exclusivity of this phenomenon to melanoma. We believe our manuscript will encourage awareness in our colleagues and stimulate interest in further studies to elucidate the mechanisms of ICI-induced vitiligo in both melanoma and non-melanoma cancers, and to understand whether this phenomenon holds the same positive prognostic value in both cancer groups. This is a retrospective cohort study from a single-institution's electronic medical record for cancer patients treated with ICIs who subsequently developed vitiligo. We identified 151 patients with ICI-induced vitiligo, 19 (12.6%) non-melanoma and 132 (77.4%) melanoma patients. Time to onset of vitiligo was nearly doubled in the non-melanoma cohort, however, this is confounded by possible delayed diagnosis or under reporting of this asymptomatic condition in patients who do not regularly receive skin exams. The majority of patients had a stable course of vitiligo with 91.4% receiving no treatment in this largely Caucasian cohort. Two patients with non-melanoma cancers and Fitzpatrick type IV or above skin received treatment with narrowband ultraviolet B light therapy and topical steroids with near-complete response. This study highlights the occurrence of ICI-induced vitiligo in a variety of non-melanoma cancers, where skin of color patients will be more prevalent and the need for treatment will potentially be more urgent. Further study is needed to elucidate the mechanism of ICI-induced vitiligo and determine if non-melanoma cancers have the same association between vitiligo and increased tumor response.
Collapse
Affiliation(s)
- Jonathan Lo
- Baylor College of Medicine, Houston, TX, USA
| | | | - Monika F Keiser
- University of Texas McGovern Medical School, Houston, TX, USA
| | - Anisha B Patel
- Department of Dermatology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
169
|
Sugiyarto G, Lau D, Hill SL, Arcia-Anaya D, Boulanger DSM, Parkes EE, James E, Elliott T. Reactivation of low avidity tumor-specific CD8 + T cells associates with immunotherapeutic efficacy of anti-PD-1. J Immunother Cancer 2023; 11:e007114. [PMID: 37586767 PMCID: PMC10432680 DOI: 10.1136/jitc-2023-007114] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2023] [Indexed: 08/18/2023] Open
Abstract
BACKGROUND CD8+ T cells are a highly diverse population of cells with distinct phenotypic functions that can influence immunotherapy outcomes. Further insights on the roles of CD8+ specificities and TCR avidity of naturally arising tumor-specific T cells, where both high and low avidity T cells recognizing the same peptide-major histocompatibility complex (pMHC) coexist in the same tumor, are crucial for understanding T cell exhaustion and resistance to PD-1 immunotherapy. METHODS CT26 models were treated with anti-PD-1 on days 3, 6 and 9 following subcutaneous tumor implantation generating variable responses during early tumor development. Tetramer staining was performed to determine the frequency and avidity of CD8+ T cells targeting the tumor-specific epitope GSW11 and confirmed with tetramer competition assays. Functional characterization of high and low avidity GSW11-specific CD8+ T cells was conducted using flow cytometry and bulk RNA-seq. In vitro cytotoxicity assays and in vivo adoptive transfer experiments were performed to determine the cytotoxicity of high and low avidity populations. RESULTS Treatment success with anti-PD-1 was associated with the preferential expansion of low avidity (Tetlo) GSW11-specific CD8+ T cells with Vβ TCR expressing clonotypes. High avidity T cells (Tethi), if present, were only found in progressing PD-1 refractory tumors. Tetlo demonstrated precursor exhausted or progenitor T cell phenotypes marked by higher expression of Tcf-1 and T-bet, and lower expression of the exhaustion markers CD39, PD-1 and Eomes compared with Tethi, whereas Tethi cells were terminally exhausted. Transcriptomics analyses showed pathways related to TCR signaling, cytotoxicity and oxidative phosphorylation were significantly enriched in Tetlo found in both regressing and progressing tumors compared with Tethi, whereas genes related to DNA damage, apoptosis and autophagy were downregulated. In vitro studies showed that Tetlo exhibits higher cytotoxicity than Tethi. Adoptive transfer of Tetlo showed more effective tumor control than Tethi, and curative responses were achieved when Tetlo was combined with two doses of anti-PD-1. CONCLUSIONS Targeting subdominant T cell responses with lower avidity against pMHC affinity neoepitopes showed potential for improving PD-1 immunotherapy. Future interventions may consider expanding low avidity populations via vaccination or adoptive transfer.
Collapse
Affiliation(s)
- Gessa Sugiyarto
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Doreen Lau
- Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Samuel Luke Hill
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, UK
| | - David Arcia-Anaya
- Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Denise S M Boulanger
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Eileen E Parkes
- Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Edward James
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Tim Elliott
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, UK
- Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
170
|
Alhumaid M, Dinakaran D, Smylie M, Walker J, Joseph K. Can radiation restore immunotherapy response in metastatic melanoma refractory to checkpoint inhibitors: An institutional experience in salvaging immunotherapy resistant disease. Radiother Oncol 2023; 185:109712. [PMID: 37178931 DOI: 10.1016/j.radonc.2023.109712] [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: 12/15/2022] [Revised: 04/29/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023]
Abstract
Melanoma is an immunogenically active tumor with abundantly expressed lymphoid infiltration. Immunotherapy(IO) has proven as a promising treatment option for melanoma but treatment resistance remains as an issue in the majority of patients.There is emerging evidence that radiotherapy (RT) could modulate the tumor microenvironment, increase antigen presentation, and augment adaptive antitumor immunity. Our objective is to evaluate overall treatment response and safety in patients with metastatic melanoma who progressed while on IO, and were treated with RT concurrently with IO for progressive sites.
Collapse
Affiliation(s)
- Mohannad Alhumaid
- Division of Radiation Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Deepak Dinakaran
- Division of Radiation Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Michael Smylie
- Division of Medical Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Alberta, Canada
| | - John Walker
- Division of Medical Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Kurian Joseph
- Division of Radiation Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
171
|
Salhi Y, De Wit R, Robbrecht D. Immunotherapy in Urothelial Cancer: Stop When Achieving a Response, Restart upon Disease Progression. Cancers (Basel) 2023; 15:3654. [PMID: 37509315 PMCID: PMC10378071 DOI: 10.3390/cancers15143654] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/13/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Since there is no clear consensus on optimal treatment duration of PD-(L)1 targeting checkpoint inhibitors in the setting of urothelial cancer (UC) patients, even patients with durable responses are often treated up to 2 years. It is questionable whether this is necessary and whether quality of life improves when treatment is discontinued earlier and restarted when necessary. METHODS We collected available data from locally advanced or metastatic UC patients within the Netherlands between September 2017 and December 2019 treated with first or second-line pembrolizumab, to evaluate treatment duration, reasons for discontinuation, subsequent treatments and survival outcomes. RESULTS Data were available from 74 patients: 85% (63/74) of patients had a treatment duration of 12 months or shorter, and in seven out of them, treatment was discontinued for another reason than progressive disease. Two patients (3%) had a treatment duration between 12 and 24 months, and eight patients (11%) completed 24 months of treatment. Survival at data cut-off (1 July 2020) with a median follow-up of 35 months was 100% in patients with partial or complete response (6/7 patients) and treatment duration ≤ 12 months, and 100% in patients treated for 24 months. In total, three patients were re-treated with pembrolizumab upon progressive disease during follow-up. CONCLUSIONS In patients who reach partial or complete response during treatment with a PD-(L)1 targeting checkpoint inhibitor, early discontinuation of treatment with pembrolizumab and restart if necessary seems to be reasonable with preserved favorable outcomes. This article should drive further efforts to optimize the treatment duration for patients who respond to treatment with pembrolizumab.
Collapse
Affiliation(s)
- Youssra Salhi
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Watermolenplein 40, 3015 GD Rotterdam, The Netherlands
| | - Ronald De Wit
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Watermolenplein 40, 3015 GD Rotterdam, The Netherlands
| | - Debbie Robbrecht
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Watermolenplein 40, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
172
|
Baruch EN, Nagarajan P, Gleber-Netto FO, Rao X, Xie T, Akhter S, Adewale A, Shajedul I, Mattson BJ, Ferrarotto R, Wong MK, Davies MA, Jindal S, Basu S, Harwood C, Leigh I, Ajami N, Futreal A, Castillo M, Gunaratne P, Goepfert RP, Khushalani N, Wang J, Watowich S, Calin GA, Migden MR, Vermeer P, D’Silva N, Yaniv D, Burks JK, Gomez J, Dougherty PM, Tsai KY, Allison JP, Sharma P, Wargo J, Myers JN, Gross ND, Amit M. Inflammation induced by tumor-associated nerves promotes resistance to anti-PD-1 therapy in cancer patients and is targetable by interleukin-6 blockade. RESEARCH SQUARE 2023:rs.3.rs-3161761. [PMID: 37503252 PMCID: PMC10371163 DOI: 10.21203/rs.3.rs-3161761/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
While the nervous system has reciprocal interactions with both cancer and the immune system, little is known about the potential role of tumor associated nerves (TANs) in modulating anti-tumoral immunity. Moreover, while peri-neural invasion is a well establish poor prognostic factor across cancer types, the mechanisms driving this clinical effect remain unknown. Here, we provide clinical and mechniastic association between TANs damage and resistance to anti-PD-1 therapy. Using electron microscopy, electrical conduction studies, and tumor samples of cutaneous squamous cell carcinoma (cSCC) patients, we showed that cancer cells can destroy myelin sheath and induce TANs degeneration. Multi-omics and spatial analyses of tumor samples from cSCC patients who underwent neoadjuvant anti-PD-1 therapy demonstrated that anti-PD-1 non-responders had higher rates of peri-neural invasion, TANs damage and degeneration compared to responders, both at baseline and following neoadjuvant treatment. Tumors from non-responders were also characterized by a sustained signaling of interferon type I (IFN-I) - known to both propagate nerve degeneration and to dampen anti-tumoral immunity. Peri-neural niches of non-responders were characterized by higher immune activity compared to responders, including immune-suppressive activity of M2 macrophages, and T regulatory cells. This tumor promoting inflammation expanded to the rest of the tumor microenvironment in non-responders. Anti-PD-1 efficacy was dampened by inducing nerve damage prior to treatment administration in a murine model. In contrast, anti-PD-1 efficacy was enhanced by denervation and by interleukin-6 blockade. These findings suggested a potential novel anti-PD-1 resistance drived by TANs damage and inflammation. This resistance mechanism is targetable and may have therapeutic implications in other neurotropic cancers with poor response to anti-PD-1 therapy such as pancreatic, prostate, and breast cancers.
Collapse
Affiliation(s)
- Erez N. Baruch
- Division of Cancer Medicine, Hematology and Oncology Fellowship program, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Frederico O. Gleber-Netto
- Department of Head and Neck Surgery, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiayu Rao
- Department of Bioinformatics and Computational Biology, Division of Basic Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tongxin Xie
- Department of Head and Neck Surgery, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shamima Akhter
- Department of Head and Neck Surgery, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Adebayo Adewale
- Department of Head and Neck Surgery, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Islam Shajedul
- Department of Head and Neck Surgery, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Brandi J Mattson
- The Neurodegeneration Consortium, Therapeutics Discovery Division, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Renata Ferrarotto
- Department of Head and Neck Thoracic Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael K. Wong
- Department of Melanoma Medical Oncology, Division of Cancer Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael A Davies
- Department of Melanoma Medical Oncology, Division of Cancer Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sonali Jindal
- Department of Immunology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sreyashi Basu
- Department of Immunology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Catherine Harwood
- Department of Dermatology, Royal London Hospital, Barts Health NHS Trust, Centre for Cell Biology and Cutaneous Research, Blizard Institute Barts and the London School of Medicine and Dentistry Queen Mary University of London, UK
| | - Irene Leigh
- Department of Dermatology, Royal London Hospital, Barts Health NHS Trust, Centre for Cell Biology and Cutaneous Research, Blizard Institute Barts and the London School of Medicine and Dentistry Queen Mary University of London, UK
| | - Nadim Ajami
- Department of Genomic Medicine, Division of Cancer Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andrew Futreal
- Department of Genomic Medicine, Division of Cancer Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Micah Castillo
- Department of Biology and Biochemistry, University of Houston Sequencing and Gene Editing Core, University of Houston, Houston, TX, USA
| | - Preethi Gunaratne
- Department of Biology and Biochemistry, University of Houston Sequencing and Gene Editing Core, University of Houston, Houston, TX, USA
| | - Ryan P. Goepfert
- Department of Head and Neck Surgery, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Jing Wang
- Department of Bioinformatics and Computational Biology, Division of Basic Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stephanie Watowich
- Department of Immunology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - George A Calin
- Department of Translational Molecular Pathology, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael R. Migden
- Department of Dermatology, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paola Vermeer
- Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD, USA
| | - Nisha D’Silva
- Department of Dentistry & Pathology, the University of Michigan, Ann Arbor, MI, USA
| | - Dan Yaniv
- Department of Head and Neck Surgery, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jared K Burks
- Department of Leukemia, Division of Cancer Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Javier Gomez
- Department of Leukemia, Division of Cancer Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Patrick M Dougherty
- Department of Pain Medicine, Division of Anesthesiology, Critical Care, and Pain Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kenneth Y. Tsai
- Department of Tumor Biology, Moffitt Cancer Center, Tampa, FL, USA
| | - James P Allison
- Department of Immunology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Padmanee Sharma
- Department of Immunology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer Wargo
- Department of Genomic Medicine, Division of Cancer Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Surgical Oncology, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jeffrey N. Myers
- Department of Head and Neck Surgery, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Neil D. Gross
- Department of Head and Neck Surgery, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Moran Amit
- Department of Head and Neck Surgery, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Genomic Medicine, Division of Cancer Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX
| |
Collapse
|
173
|
Martinis E, Ricci C, Trevisan C, Tomadini G, Tonon S. Cancer Vaccines: From the State of the Art to the Most Promising Frontiers in the Treatment of Colorectal Cancer. Pharmaceutics 2023; 15:1969. [PMID: 37514155 PMCID: PMC10383643 DOI: 10.3390/pharmaceutics15071969] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/06/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Colorectal cancer represents 10% of all new cancer cases each year and accounts for almost 10% of all cancer deaths. According to the WHO, by 2040 there will be a 60% increase in colorectal cancer cases. These data highlight the need to explore new therapeutic strategies. Classical interventions include surgical resection, chemotherapy and radiotherapy, which are invasive strategies that have many side effects on the patients and greatly affect their quality of life. A great advance in the treatment of this cancer type, as well as of all the others, could be the development of a vaccination strategy preventing the onset, the progression or the relapse of the pathology. In this review, we summarize the main vaccination strategies that are being studied for the treatment of colorectal cancer (CRC) and finally explore the possibility of using B-cells for the development of a new type of vaccine.
Collapse
Affiliation(s)
- Eleonora Martinis
- Department of Medicine, University of Udine, Piazzale Kolbe 4 Udine, 33100 Udine, Italy
| | - Carolina Ricci
- Department of Medicine, University of Udine, Piazzale Kolbe 4 Udine, 33100 Udine, Italy
| | - Caterina Trevisan
- Department of Medicine, University of Udine, Piazzale Kolbe 4 Udine, 33100 Udine, Italy
| | - Gaia Tomadini
- Department of Medicine, University of Udine, Piazzale Kolbe 4 Udine, 33100 Udine, Italy
| | - Silvia Tonon
- Department of Medicine, University of Udine, Piazzale Kolbe 4 Udine, 33100 Udine, Italy
| |
Collapse
|
174
|
Pal SK, Albiges L, Tomczak P, Suárez C, Voss MH, de Velasco G, Chahoud J, Mochalova A, Procopio G, Mahammedi H, Zengerling F, Kim C, Osawa T, Angel M, Gupta S, Khan O, Bergthold G, Liu B, Kalaitzidou M, Huseni M, Scheffold C, Powles T, Choueiri TK. Atezolizumab plus cabozantinib versus cabozantinib monotherapy for patients with renal cell carcinoma after progression with previous immune checkpoint inhibitor treatment (CONTACT-03): a multicentre, randomised, open-label, phase 3 trial. Lancet 2023; 402:185-195. [PMID: 37290461 PMCID: PMC11017728 DOI: 10.1016/s0140-6736(23)00922-4] [Citation(s) in RCA: 97] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 04/25/2023] [Accepted: 04/28/2023] [Indexed: 06/10/2023]
Abstract
BACKGROUND Immune checkpoint inhibitors are the standard of care for first-line treatment of patients with metastatic renal cell carcinoma, yet optimised treatment of patients whose disease progresses after these therapies is unknown. The aim of this study was to determine whether adding atezolizumab to cabozantinib delayed disease progression and prolonged survival in patients with disease progression on or after previous immune checkpoint inhibitor treatment. METHODS CONTACT-03 was a multicentre, randomised, open-label, phase 3 trial, done in 135 study sites in 15 countries in Asia, Europe, North America, and South America. Patients aged 18 years or older with locally advanced or metastatic renal cell carcinoma whose disease had progressed with immune checkpoint inhibitors were randomly assigned (1:1) to receive atezolizumab (1200 mg intravenously every 3 weeks) plus cabozantinib (60 mg orally once daily) or cabozantinib alone. Randomisation was done through an interactive voice-response or web-response system in permuted blocks (block size four) and stratified by International Metastatic Renal Cell Carcinoma Database Consortium risk group, line of previous immune checkpoint inhibitor therapy, and renal cell carcinoma histology. The two primary endpoints were progression-free survival per blinded independent central review and overall survival. The primary endpoints were assessed in the intention-to-treat population and safety was assessed in all patients who received at least one dose of study drug. The trial is registered with ClinicalTrials.gov, NCT04338269, and is closed to further accrual. FINDINGS From July 28, 2020, to Dec 27, 2021, 692 patients were screened for eligibility, 522 of whom were assigned to receive atezolizumab-cabozantinib (263 patients) or cabozantinib (259 patients). 401 (77%) patients were male and 121 (23%) patients were female. At data cutoff (Jan 3, 2023), median follow-up was 15·2 months (IQR 10·7-19·3). 171 (65%) patients receiving atezolizumab-cabozantinib and 166 (64%) patients receiving cabozantinib had disease progression per central review or died. Median progression-free survival was 10·6 months (95% CI 9·8-12·3) with atezolizumab-cabozantinib and 10·8 months (10·0-12·5) with cabozantinib (hazard ratio [HR] for disease progression or death 1·03 [95% CI 0·83-1·28]; p=0·78). 89 (34%) patients in the atezolizumab-cabozantinib group and 87 (34%) in the cabozantinib group died. Median overall survival was 25·7 months (95% CI 21·5-not evaluable) with atezolizumab-cabozantinib and was not evaluable (21·1-not evaluable) with cabozantinib (HR for death 0·94 [95% CI 0·70-1·27]; p=0·69). Serious adverse events occurred in 126 (48%) of 262 patients treated with atezolizumab-cabozantinib and 84 (33%) of 256 patients treated with cabozantinib; adverse events leading to death occurred in 17 (6%) patients in the atezolizumab-cabozantinib group and nine (4%) in the cabozantinib group. INTERPRETATION The addition of atezolizumab to cabozantinib did not improve clinical outcomes and led to increased toxicity. These results should discourage sequential use of immune checkpoint inhibitors in patients with renal cell carcinoma outside of clinical trials. FUNDING F Hoffmann-La Roche and Exelixis.
Collapse
Affiliation(s)
- Sumanta Kumar Pal
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Laurence Albiges
- Department of Cancer Medicine, Institut Gustave Roussy, Villejuif, France
| | - Piotr Tomczak
- Department of Oncology, Poznan University of Medical Sciences, Poznan, Poland
| | - Cristina Suárez
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Martin H Voss
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Jad Chahoud
- Department of Genitourinary Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Anastasia Mochalova
- Department of Chemotherapy, Clinical Hospital Number 1 MEDSI, Moscow, Russia
| | - Giuseppe Procopio
- Department of Medical Oncology, Fondazione Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Hakim Mahammedi
- Department of Medical Oncology, Jean Perrin Center, Clermont-Ferrand, France
| | | | - Chan Kim
- Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Takahiro Osawa
- Department of Urology, Hokkaido University Hospital, Sapporo, Japan
| | - Martín Angel
- Department of Genitourinary Cancer, Instituto Alexander Fleming, Buenos Aires, Argentina
| | | | | | | | - Bo Liu
- Genentech, South San Francisco, CA, USA
| | | | | | | | - Thomas Powles
- Barts Cancer Institute, Centre for Experimental Cancer Medicine, Queen Mary University of London, London, UK
| | - Toni K Choueiri
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
175
|
Olateju OA, Zeng Z, Thornton JD, Mgbere O, Essien EJ. Management of metastatic melanoma in Texas: disparities in the utilization of immunotherapy following the regulatory approval of immune checkpoint inhibitors. BMC Cancer 2023; 23:655. [PMID: 37442992 DOI: 10.1186/s12885-023-11142-4] [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: 12/29/2022] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND The utilization of modern-immunotherapies, notably immune checkpoint inhibitors (ICIs), has increased markedly in patients with metastatic melanoma over the past decade and are recommended as standard treatment. Given their increasing adoption in routine care for melanoma, understanding patient access to immunotherapy and patterns of its use in Texas is crucial as it remains one of the few states without Medicaid expansion and with high rates of the uninsured population. The objectives of this study were to examine the trend in the utilization of immunotherapy and to determine factors associated with immunotherapy utilization among patients with metastatic melanoma in the era of ICIs in Texas. METHODS A retrospective cohort study was conducted using the Texas Cancer Registry (TCR) database. The cohort comprised of adult (≥ 18 years) patients with metastatic melanoma diagnosed between June 2011 and December 2018. The trend in immunotherapy utilization was assessed by determining the proportion of patients receiving immunotherapy each year. The Average Annual Percent Change (AAPC) in immunotherapy utilization was assessed using joinpoint regression, while multivariable logistic regression was used to determine the association between patient characteristics and immunotherapy receipt. RESULTS A total of 1,795 adult patients with metastatic melanoma were identified from the TCR. Immunotherapy utilization was higher among younger patients, those with no comorbidities, and patients with private insurance. Multivariable analysis showed that the likelihood of receipt of immunotherapy decreased with older age [(adjusted Odds Ratio (aOR), 0.92; 95% CI, 0.89- 0.93, p = 0.001], living in high poverty neighborhood (aOR, 0.52; 95% CI, 0.44 - 0.66, p < 0.0001), having Medicaid (aOR, 0.58; 95% CI, 0.44 - 0.73, p = 0.02), being uninsured (aOR, 0.49; 95% CI, 0.31 - 0.64, p = 0.01), and having comorbidities (CCI score 1: aOR, 0.48; 95% CI, 0.34 - 0.71, p = 0.003; CCI score ≥ 2: aOR, 0.32; 95% CI, 0.16 - 0.56, p < 0.0001). CONCLUSIONS AND RELEVANCE This cohort study identified sociodemographic and socioeconomic disparities in access to immunotherapy in Texas, highlighting the need for policies such as Medicaid expansion that would increase equitable access to this innovative therapy.
Collapse
Affiliation(s)
- Olajumoke A Olateju
- Department of Pharmaceutical Health Outcomes and Policy, College of Pharmacy, University of Houston, Houston, TX, USA
| | - Zhen Zeng
- Department of Pharmaceutical Health Outcomes and Policy, College of Pharmacy, University of Houston, Houston, TX, USA
| | - J Douglas Thornton
- Department of Pharmaceutical Health Outcomes and Policy, College of Pharmacy, University of Houston, Houston, TX, USA
| | - Osaro Mgbere
- Department of Pharmaceutical Health Outcomes and Policy, College of Pharmacy, University of Houston, Houston, TX, USA
- Institute of Community Health, University of Houston College of Pharmacy, Houston, TX, USA
- Public Health Science and Surveillance Division, Houston Health Department, Houston, TX, USA
| | - Ekere James Essien
- Department of Pharmaceutical Health Outcomes and Policy, College of Pharmacy, University of Houston, Houston, TX, USA.
- Institute of Community Health, University of Houston College of Pharmacy, Houston, TX, USA.
| |
Collapse
|
176
|
Nardin C, Hennemann A, Diallo K, Funck-Brentano E, Puzenat E, Heidelberger V, Jeudy G, Samimi M, Lesage C, Boussemart L, Peuvrel L, Rouanet J, Brunet-Possenti F, Gerard E, Seris A, Jouary T, Saint-Jean M, Puyraveau M, Saiag P, Aubin F. Efficacy of Immune Checkpoint Inhibitor (ICI) Rechallenge in Advanced Melanoma Patients' Responders to a First Course of ICI: A Multicenter National Retrospective Study of the French Group of Skin Cancers (Groupe de Cancérologie Cutanée, GCC). Cancers (Basel) 2023; 15:3564. [PMID: 37509227 PMCID: PMC10377277 DOI: 10.3390/cancers15143564] [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: 04/24/2023] [Revised: 06/07/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND The long-term effectiveness of immune checkpoint inhibitor (ICI) rechallenge for progressive or recurrent advanced melanoma following previous disease control induced by ICI has not been thoroughly described in the literature. PATIENTS AND METHODS In this retrospective multicenter national real-life study, we enrolled patients who had been rechallenged with an ICI after achieving disease control with a first course of ICI, which was subsequently interrupted. The primary objective was to evaluate tumor response, while the secondary objectives included assessing the safety profile, identifying factors associated with tumor response, and evaluating survival outcomes. RESULTS A total of 85 patients from 12 centers were included in the study. These patients had advanced (unresectable stage III or stage IV) melanoma that had been previously treated and controlled with a first course of ICI before undergoing rechallenge with ICI. The rechallenge treatments consisted of pembrolizumab (n = 44, 52%), nivolumab (n = 35, 41%), ipilimumab (n = 2, 2%), or ipilimumab plus nivolumab (n = 4, 5%). The best overall response rate was 54%. The best response was a complete response in 30 patients (35%), a partial response in 16 patients (19%), stable disease in 18 patients (21%) and progressive disease in 21 patients (25%). Twenty-eight adverse events (AEs) were reported in 23 patients (27%), including 18 grade 1-2 AEs in 14 patients (16%) and 10 grade 3-4 AEs in nine patients (11%). The median progression-free survival (PFS) was 21 months, and the median overall survival (OS) was not reached at the time of analysis. Patients who received another systemic treatment (chemotherapy, targeted therapy or clinical trial) between the two courses of ICI had a lower response to rechallenge (p = 0.035) and shorter PFS (p = 0.016). CONCLUSION Rechallenging advanced melanoma patients with ICI after previous disease control induced by these inhibitors resulted in high response rates (54%) and disease control (75%). Therefore, ICI rechallenge should be considered as a relevant therapeutic option.
Collapse
Affiliation(s)
- Charlée Nardin
- Service de Dermatologie, Centre Hospitalier Universitaire, 25000 Besancon, France
- Université Franche Comté, Inserm 1098 RIGHT, 25020 Besancon, France
| | - Aymeric Hennemann
- Service de Dermatologie, Centre Hospitalier Universitaire, 25000 Besancon, France
| | - Kadiatou Diallo
- Centre de Méthodologie Clinique, Centre Hospitalier Universitaire, 25030 Besancon, France
| | - Elisa Funck-Brentano
- Université Paris-Saclay, UVSQ, EA4340-BECCOH, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Ambroise-Paré, Service de Dermatologie Générale et Oncologique, 92104 Boulogne-Billancourt, France
| | - Eve Puzenat
- Service de Dermatologie, Centre Hospitalier Universitaire, 25000 Besancon, France
| | | | - Géraldine Jeudy
- Service de Dermatologie, Centre Hospitalier Universitaire, Hôpital Le Bocage, 21079 Dijon, France
| | - Mahtab Samimi
- Service de Dermatologie, Centre Hospitalier Universitaire, BIP 1282, INRA-Université de Tours, 37020 Tours, France
| | - Candice Lesage
- Service de Dermatologie, Centre Hospitalier Universitaire, 34295 Montpellier, France
| | - Lise Boussemart
- Service de Dermatologie, Centre Hospitalier Universitaire, Université de Nantes, INSERM, Immunology and New Concepts in Immunotherapy, INCIT, UMR 1302, 44000 Nantes, France
| | - Lucie Peuvrel
- Institut de Cancérologie de l'Ouest, 44800 Saint-Herblain, France
| | - Jacques Rouanet
- Service de Dermatologie, Centre Hospitalier Universitaire, 63003 Clermont-Ferrand, France
| | | | - Emilie Gerard
- Service de Dermatologie, Centre Hospitalier Universitaire, 33075 Bordeaux, France
| | - Alice Seris
- Oncologie Médicale, Centre Hospitalier, 64046 Pau, France
| | - Thomas Jouary
- Oncologie Médicale, Centre Hospitalier, 64046 Pau, France
| | | | - Marc Puyraveau
- Centre de Méthodologie Clinique, Centre Hospitalier Universitaire, 25030 Besancon, France
| | - Philippe Saiag
- Université Paris-Saclay, UVSQ, EA4340-BECCOH, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Ambroise-Paré, Service de Dermatologie Générale et Oncologique, 92104 Boulogne-Billancourt, France
| | - François Aubin
- Service de Dermatologie, Centre Hospitalier Universitaire, 25000 Besancon, France
- Université Franche Comté, Inserm 1098 RIGHT, 25020 Besancon, France
| |
Collapse
|
177
|
Ho P, Melms JC, Rogava M, Frangieh CJ, Poźniak J, Shah SB, Walsh Z, Kyrysyuk O, Amin AD, Caprio L, Fullerton BT, Soni RK, Ager CR, Biermann J, Wang Y, Khosravi-Maharlooei M, Zanetti G, Mu M, Fatima H, Moore EK, Vasan N, Bakhoum SF, Reiner SL, Bernatchez C, Sykes M, Mace EM, Wucherpfennig KW, Schadendorf D, Bechter O, Shah P, Schwartz GK, Marine JC, Izar B. The CD58-CD2 axis is co-regulated with PD-L1 via CMTM6 and shapes anti-tumor immunity. Cancer Cell 2023; 41:1207-1221.e12. [PMID: 37327789 PMCID: PMC10524902 DOI: 10.1016/j.ccell.2023.05.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/10/2023] [Accepted: 05/22/2023] [Indexed: 06/18/2023]
Abstract
The cell-autonomous balance of immune-inhibitory and -stimulatory signals is a critical process in cancer immune evasion. Using patient-derived co-cultures, humanized mouse models, and single-cell RNA-sequencing of patient melanomas biopsied before and on immune checkpoint blockade, we find that intact cancer cell-intrinsic expression of CD58 and ligation to CD2 is required for anti-tumor immunity and is predictive of treatment response. Defects in this axis promote immune evasion through diminished T cell activation, impaired intratumoral T cell infiltration and proliferation, and concurrently increased PD-L1 protein stabilization. Through CRISPR-Cas9 and proteomics screens, we identify and validate CMTM6 as critical for CD58 stability and upregulation of PD-L1 upon CD58 loss. Competition between CD58 and PD-L1 for CMTM6 binding determines their rate of endosomal recycling over lysosomal degradation. Overall, we describe an underappreciated yet critical axis of cancer immunity and provide a molecular basis for how cancer cells balance immune inhibitory and stimulatory cues.
Collapse
Affiliation(s)
- Patricia Ho
- Department of Medicine, Division of Hematology and Oncology, Columbia University, New York, NY 10032, USA; Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Johannes C Melms
- Department of Medicine, Division of Hematology and Oncology, Columbia University, New York, NY 10032, USA; Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Meri Rogava
- Department of Medicine, Division of Hematology and Oncology, Columbia University, New York, NY 10032, USA; Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Chris J Frangieh
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Klarman Cell Observatory, the Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Joanna Poźniak
- Laboratory for Molecular Cancer Biology, Center for Cancer Biology, VIB, 3000 Leuven, Belgium; Department of Oncology, KU Leuven, 3000 Leuven, Belgium
| | - Shivem B Shah
- Department of Medicine, Division of Hematology and Oncology, Columbia University, New York, NY 10032, USA; Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Zachary Walsh
- Department of Medicine, Division of Hematology and Oncology, Columbia University, New York, NY 10032, USA; Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Oleksandr Kyrysyuk
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Amit Dipak Amin
- Department of Medicine, Division of Hematology and Oncology, Columbia University, New York, NY 10032, USA; Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Lindsay Caprio
- Department of Medicine, Division of Hematology and Oncology, Columbia University, New York, NY 10032, USA; Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Benjamin T Fullerton
- Department of Medicine, Division of Hematology and Oncology, Columbia University, New York, NY 10032, USA
| | - Rajesh Kumar Soni
- Proteomics and Macromolecular Crystallography Shared Resource, Columbia University, New York, NY 10032, USA
| | - Casey R Ager
- Department of Medicine, Division of Hematology and Oncology, Columbia University, New York, NY 10032, USA; Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Jana Biermann
- Department of Medicine, Division of Hematology and Oncology, Columbia University, New York, NY 10032, USA; Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA; Program for Mathematical Genomics, Department of Systems Biology, Columbia University, New York, NY 10032, USA
| | - Yiping Wang
- Department of Medicine, Division of Hematology and Oncology, Columbia University, New York, NY 10032, USA; Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA; Program for Mathematical Genomics, Department of Systems Biology, Columbia University, New York, NY 10032, USA
| | - Mohsen Khosravi-Maharlooei
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA; Department of Immunology, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Giorgia Zanetti
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Michael Mu
- Department of Medicine, Division of Hematology and Oncology, Columbia University, New York, NY 10032, USA; Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Hijab Fatima
- Department of Pediatrics, Columbia University, New York, NY 10032, USA
| | - Emily K Moore
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA; Department of Medicine, Division of Rheumatology, Columbia University, New York, NY 10032, USA
| | - Neil Vasan
- Department of Medicine, Division of Hematology and Oncology, Columbia University, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA
| | - Samuel F Bakhoum
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Steven L Reiner
- Department of Pediatrics, Columbia University, New York, NY 10032, USA; Department of Microbiology and Immunology, Columbia University, New York, NY 10032, USA
| | - Chantale Bernatchez
- Department of Medical Oncology, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Megan Sykes
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA; Department of Microbiology and Immunology, Columbia University, New York, NY 10032, USA; Department of Surgery, Columbia University, New York, NY 10032, USA
| | - Emily M Mace
- Department of Pediatrics, Columbia University, New York, NY 10032, USA
| | - Kai W Wucherpfennig
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen and German Cancer Consortium, Partner Site, 45147 Essen, Germany
| | | | - Parin Shah
- Department of Medicine, Division of Hematology and Oncology, Columbia University, New York, NY 10032, USA
| | - Gary K Schwartz
- Department of Medicine, Division of Hematology and Oncology, Columbia University, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA
| | - Jean-Christophe Marine
- Laboratory for Molecular Cancer Biology, Center for Cancer Biology, VIB, 3000 Leuven, Belgium; Department of Oncology, KU Leuven, 3000 Leuven, Belgium
| | - Benjamin Izar
- Department of Medicine, Division of Hematology and Oncology, Columbia University, New York, NY 10032, USA; Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA; Program for Mathematical Genomics, Department of Systems Biology, Columbia University, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
178
|
Ozbay Kurt FG, Lasser S, Arkhypov I, Utikal J, Umansky V. Enhancing immunotherapy response in melanoma: myeloid-derived suppressor cells as a therapeutic target. J Clin Invest 2023; 133:e170762. [PMID: 37395271 DOI: 10.1172/jci170762] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2023] Open
Abstract
Despite the remarkable success of immune checkpoint inhibitors (ICIs) in melanoma treatment, resistance to them remains a substantial clinical challenge. Myeloid-derived suppressor cells (MDSCs) represent a heterogeneous population of myeloid cells that can suppress antitumor immune responses mediated by T and natural killer cells and promote tumor growth. They are major contributors to ICI resistance and play a crucial role in creating an immunosuppressive tumor microenvironment. Therefore, targeting MDSCs is considered a promising strategy to improve the therapeutic efficacy of ICIs. This Review describes the mechanism of MDSC-mediated immune suppression, preclinical and clinical studies on MDSC targeting, and potential strategies for inhibiting MDSC functions to improve melanoma immunotherapy.
Collapse
Affiliation(s)
- Feyza Gul Ozbay Kurt
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | - Samantha Lasser
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | - Ihor Arkhypov
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Viktor Umansky
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| |
Collapse
|
179
|
Ayati N, Jamshidi-Araghi Z, Hoellwerth M, Schweighofer-Zwink G, Hitzl W, Koelblinger P, Pirich C, Beheshti M. Predictive value and accuracy of [ 18F]FDG PET/CT modified response criteria for checkpoint immunotherapy in patients with advanced melanoma. Eur J Nucl Med Mol Imaging 2023; 50:2715-2726. [PMID: 37140669 PMCID: PMC10317870 DOI: 10.1007/s00259-023-06247-8] [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: 01/30/2023] [Accepted: 04/24/2023] [Indexed: 05/05/2023]
Abstract
PURPOSE Immune checkpoint inhibitors (ICIs) are widely used in metastatic melanoma and dramatically alter the treatment of these patients. Given the high cost and potential toxicity, a reliable method for evaluating treatment response is needed. In this study, we assessed tumor response in patients with metastatic melanoma treated with ICIs using three modified response criteria: PET Response Evaluation Criteria for Immunotherapy (PERCIMT), PET Response Criteria in Solid Tumors for up to Five Lesions (PERCIST5), and immunotherapy-modified PET Response Criteria in Solid Tumors for up to Five Lesions (imPERCIST5). METHODS Ninety-one patients with non-resectable stage IV metastatic melanoma who received ICIs were retrospectively enrolled in this study. Each patient had two [18F]FDG PET/CT scans performed before and after ICI therapy. Responses at the follow-up scan were evaluated according to PERCIMT, PERCIST5, and imPERCIST5 criteria. Patients were classified into four groups: complete metabolic response (CMR), partial metabolic response (PMR), progressive metabolic disease (PMD), and stable metabolic disease (SMD). To assess the "disease control rate," two groups have been defined based on each criterion: patients with CMR, PMR, and SMD as "disease-controlled group (i.e., responders)" and PMD as the "uncontrolled-disease group (i.e., non-responders)". The correspondence between metabolic tumor response defined by these criteria and clinical outcome was assessed and compared. RESULTS The response and the disease control rates were 40.7% and 71.4%, 41.8% and 50.5%, and 54.9% and 74.7% based on the PERCIMT, PERCIST5, and imPERCIST5 criteria, respectively. PERCIMT and imPERCIST5 showed significantly different disease control rates from that of PERCIST5 (P < 0.001), whereas it was not significant between PERCIMT and imPERCIST5. Overall survival was significantly longer in the metabolic responder groups than in the non-responder groups based on PERCIMT and PERCIST5 criteria (PERCIMT: 2.48 versus 1.47 years, P = 0.003; PERCIST5: 2.57 versus 1.81 years. P = 0.017). However, according to imPERCIST5 criterion, this difference was not observed (P = 0.12). CONCLUSION Although the appearance of new lesions can be secondary to an inflammatory response to ICIs and indicative of pseudoprogression, given the higher rate of true progression, the appearance of new lesions should be interpreted deliberately. Of the three assessed modified criteria, PERCIMT appear to provide more reliable metabolic response assessment that correlates strongly with overall patient survival.
Collapse
Affiliation(s)
- Narjess Ayati
- Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Zahra Jamshidi-Araghi
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, Muellner Hauptstrasse 48, 5020, Salzburg, Austria
- Department of Nuclear Medicine, Shahid Rajaie Cardiovascular, Medical & Research Center, Tehran, Iran
| | - Magdalena Hoellwerth
- Department of Dermatology, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Gregor Schweighofer-Zwink
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, Muellner Hauptstrasse 48, 5020, Salzburg, Austria
| | - Wolfgang Hitzl
- Biostatistics and Publication of Clinical Trial Studies, Research and Innovation Management (RIM), Paracelsus Medical University, Salzburg, Austria
- Department of Ophthalmology and Optometry, Paracelsus Medical University, Salzburg, Austria
- Research Program Experimental Ophthalmology & Glaucoma Research, Paracelsus Medical University, Salzburg, Austria
| | - Peter Koelblinger
- Department of Dermatology, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Christian Pirich
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, Muellner Hauptstrasse 48, 5020, Salzburg, Austria
| | - Mohsen Beheshti
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, Muellner Hauptstrasse 48, 5020, Salzburg, Austria.
| |
Collapse
|
180
|
Rozeman EA, Versluis JM, Sikorska K, Hoefsmit EP, Dimitriadis P, Rao D, Lacroix R, Grijpink-Ongering LG, Lopez-Yurda M, Heeres BC, van de Wiel BA, Flohil C, Sari A, Heijmink SWTPJ, van den Broek D, Broeks A, de Groot JWB, Vollebergh MA, Wilgenhof S, van Thienen JV, Haanen JBAG, Blank CU. IMPemBra: a phase 2 study comparing pembrolizumab with intermittent/short-term dual MAPK pathway inhibition plus pembrolizumab in patients with melanoma harboring the BRAFV600 mutation. J Immunother Cancer 2023; 11:e006821. [PMID: 37479483 PMCID: PMC10364170 DOI: 10.1136/jitc-2023-006821] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2023] [Indexed: 07/23/2023] Open
Abstract
BACKGROUND Continuous combination of MAPK pathway inhibition (MAPKi) and anti-programmed death-(ligand) 1 (PD-(L)1) showed high response rates, but only limited improvement in progression-free survival (PFS) at the cost of a high frequency of treatment-related adverse events (TRAE) in patients with BRAFV600-mutated melanoma. Short-term MAPKi induces T-cell infiltration in patients and is synergistic with anti-programmed death-1 (PD-1) in a preclinical melanoma mouse model. The aim of this phase 2b trial was to identify an optimal regimen of short-term MAPKi with dabrafenib plus trametinib in combination with pembrolizumab. METHODS Patients with treatment-naïve BRAFV600E/K-mutant advanced melanoma started pembrolizumab 200 mg every 3 weeks. In week 6, patients were randomized to continue pembrolizumab only (cohort 1), or to receive, in addition, intermittent dabrafenib 150 mg two times per day plus trametinib 2 mg one time per day for two cycles of 1 week (cohort 2), two cycles of 2 weeks (cohort 3), or continuously for 6 weeks (cohort 4). All cohorts continued pembrolizumab for up to 2 years. Primary endpoints were safety and treatment-adherence. Secondary endpoints were objective response rate (ORR) at week 6, 12, 18 and PFS. RESULTS Between June 2016 and August 2018, 33 patients with advanced melanoma have been included and 32 were randomized. Grade 3-4 TRAE were observed in 12%, 12%, 50%, and 63% of patients in cohort 1, 2, 3, and 4, respectively. All planned targeted therapy was given in 88%, 63%, and 38% of patients in cohort 2, 3, and 4. ORR at week 6, 12, and 18 were 38%, 63%, and 63% in cohort 1; 25%, 63%, and 75% in cohort 2; 25%, 50%, and 75% in cohort 3; and 0%, 63%, and 50% in cohort 4. After a median follow-up of 43.5 months, median PFS was 10.6 months for pembrolizumab monotherapy and not reached for patients treated with pembrolizumab and intermittent dabrafenib and trametinib (p=0.17). The 2-year and 3-year landmark PFS were both 25% for cohort 1, both 63% for cohort 2, 50% and 38% for cohort 3 and 75% and 60% for cohort 4. CONCLUSIONS The combination of pembrolizumab plus intermittent dabrafenib and trametinib seems more feasible and tolerable than continuous triple therapy. The efficacy is promising and appears to be favorable over pembrolizumab monotherapy. TRIAL REGISTRATION NUMBER NCT02625337.
Collapse
Affiliation(s)
- Elisa A Rozeman
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Judith M Versluis
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Karolina Sikorska
- Department of Biometrics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Esmée P Hoefsmit
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Petros Dimitriadis
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Disha Rao
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ruben Lacroix
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Marta Lopez-Yurda
- Department of Biometrics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Birthe C Heeres
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Bart A van de Wiel
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Claudie Flohil
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Aysegul Sari
- Department of Biometrics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Daan van den Broek
- Department of Laboratory Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Annegien Broeks
- Core Facility and Biobanking, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Marieke A Vollebergh
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Sofie Wilgenhof
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Johannes V van Thienen
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - John B A G Haanen
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Christian U Blank
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Medical Oncology, Leiden University Medical center, Leiden, The Netherlands
| |
Collapse
|
181
|
de Barros E Silva MJ, Teixeira MR, Lobo MDM, Molina AS, Bertolli E, Santos Filho IDDAO, Castro Ribeiro HS, Pelizon ACDA, Lopes Pinto CA, Duprat Neto JP. Local therapy in advanced melanoma after immune checkpoint inhibitors aiming to achieve complete response. J Cancer Res Ther 2023; 19:1272-1278. [PMID: 37787295 DOI: 10.4103/jcrt.jcrt_1684_21] [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] [Indexed: 10/04/2023]
Abstract
Background and Objectives New scenarios for local therapy have arisen after starting immune checkpoint inhibitors (ICIs) to treat advanced melanoma (AM). The aim of this study is to examine the role of local therapies with curative intention for patients with AM that have been on ICI. Methods This was a single institution, retrospective analysis of unresectable stage III or IV melanoma patients on treatment with anti-PD1 ± anti-CTLA-4 who underwent local therapy with curative intention with no other remaining sites of disease (NRD). Results Of the 170 patients treated with ICI, 19 (11.2%) met the criteria of curative intention. The median time on ICI before local therapy was 16.6 months (range: 0.92-43.2). At the time of the local treatment, the disease was controlled in 16 (84.25%) and progressing in 3 patients (15.75%); 14 patients (73.7%) treated a single lesion and 5 (26.3%) treated 2 to 3 lesions. In a median follow-up of 17 months (range: 1.51-38.2) after the local therapy and 9.8 months after the last ICI cycle (range: 0.56-31), only 2 (10.5%) out of 19 patients relapsed. Conclusions Patients with AM on treatment with ICI were able to achieve NRD after local treatment and may benefit from long-term disease control without systemic treatment.
Collapse
Affiliation(s)
| | | | | | - André Sapata Molina
- Department of Surgical Oncology, AC Camargo Cancer Center, São Paulo, Brazil
| | - Eduardo Bertolli
- Department of Surgical Oncology, AC Camargo Cancer Center, São Paulo, Brazil
| | | | | | | | | | | |
Collapse
|
182
|
Galus Ł, Michalak M, Lorenz M, Stoińska-Swiniarek R, Tusień Małecka D, Galus A, Kolenda T, Leporowska E, Mackiewicz J. Vitamin D supplementation increases objective response rate and prolongs progression-free time in patients with advanced melanoma undergoing anti-PD-1 therapy. Cancer 2023; 129:2047-2055. [PMID: 37089083 DOI: 10.1002/cncr.34718] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/28/2023] [Accepted: 01/31/2023] [Indexed: 04/25/2023]
Abstract
BACKGROUND Vitamin D3 is a prohormone with pleiotropic effects, including modulating the functions of the immune system and may affect the effectiveness of anti-PD-1 treatment in patients with cancer. According to the literature, the potential mechanism of vitamin D's influence on the effectiveness of therapy is most likely related to the amount and activity of tumor-infiltrating lymphocytes. There are data showing the effect of vitamin D on cells regulating the activity of CD8 lymphocytes. METHODS A total of 200 patients with advanced melanoma were included in the study. All patients received anti-PD-1 immunotherapy (nivolumab or pembrolizumab) as first-line treatment. Serum vitamin D levels were measured in patients both before and every 12 weeks during treatment. Part of the group had vitamin D measured retrospectively from the preserved serum. The other part of the supplementation group was tested prospectively. RESULTS The response rate in the group with low vitamin D levels and not supplemented was 36.2%, whereas in the group with normal baseline levels or a normal level obtained with supplementation was 56.0% (p = .01). Moreover, progression-free survival in these groups was 5.75 and 11.25 months, respectively (p = .03). In terms of overall survival, there was also a difference in favor of the group with normal vitamin D levels (27 vs. 31.5 months, respectively; p = .39). CONCLUSIONS In our opinion, maintaining the vitamin D level within the normal range during anti-PD-1 immunotherapy in advanced melanoma patients should be a standard procedure allowing the improvement of treatment outcomes.
Collapse
Affiliation(s)
- Łukasz Galus
- Department of Medical and Experimental Oncology, Institute of Oncology, Poznan University of Medical Sciences, Poznań, Poland
| | - Michał Michalak
- Department of Computer Science and Statistics, Poznan University of Medical Sciences, Poznań, Poland
| | - Mateusz Lorenz
- Department of Diagnostic Imaging, Heliodor Swiecicki Clinical Hospital, Poznan University of Medical Sciences, Poznań, Poland
| | - Renata Stoińska-Swiniarek
- Department of Medical and Experimental Oncology, Heliodor Swiecicki Clinical Hospital, Poznan University of Medical Sciences, Poznań, Poland
| | - Daria Tusień Małecka
- Department of Medical and Experimental Oncology, Institute of Oncology, Poznan University of Medical Sciences, Poznań, Poland
| | - Agnieszka Galus
- Department of Medical and Experimental Oncology, Heliodor Swiecicki Clinical Hospital, Poznan University of Medical Sciences, Poznań, Poland
| | - Tomasz Kolenda
- Laboratory of Cancer Genetics, Greater Poland Cancer Centre, Poznań, Poland
| | - Ewa Leporowska
- Department of Laboratory Diagnostics, Greater Poland Cancer Centre, Poznań, Poland
| | - Jacek Mackiewicz
- Department of Medical and Experimental Oncology, Institute of Oncology, Poznan University of Medical Sciences, Poznań, Poland
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, Poznań, Poland
| |
Collapse
|
183
|
Giovannini E, Bonasoni MP, D'Aleo M, Tamagnini I, Tudini M, Fais P, Pelotti S. Pembrolizumab-Induced Fatal Myasthenia, Myocarditis, and Myositis in a Patient with Metastatic Melanoma: Autopsy, Histological, and Immunohistochemical Findings-A Case Report and Literature Review. Int J Mol Sci 2023; 24:10919. [PMID: 37446095 DOI: 10.3390/ijms241310919] [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: 05/25/2023] [Revised: 06/15/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) represent a major advance in cancer treatment. The lowered immune tolerance induced by ICIs brought to light a series of immune-related adverse events (irAEs). Pembrolizumab belongs to the ICI class and is a humanized IgG4 anti-PD-1 antibody that blocks the interaction between PD-1 and PD-L1. The ICI-related irAEs involving various organ systems and myocarditis are uncommon (incidence of 0.04% to 1.14%), but they are associated with a high reported mortality. Unlike idiopathic inflammatory myositis, ICI-related myositis has been reported to frequently co-occur with myocarditis. The triad of myasthenia, myositis, and myocarditis must not be underestimated as they can rapidly deteriorate, leading to death. Herein we report a case of a patient with metastatic melanoma who fatally developed myasthenia gravis, myocarditis, and myositis, after a single cycle of pembrolizumab. Considering evidence from the literature review, autopsy, histological, and immunohistochemical investigations on heart and skeletal muscle are presented and discussed, also from a medical-legal perspective.
Collapse
Affiliation(s)
- Elena Giovannini
- Unit of Legal Medicine, Department of Medical and Surgical Sciences, University of Bologna, Via Irnerio 49, 40126 Bologna, Italy
| | - Maria Paola Bonasoni
- Pathology Unit, Azienda USL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Michele D'Aleo
- Unit of Legal Medicine, Department of Medical and Surgical Sciences, University of Bologna, Via Irnerio 49, 40126 Bologna, Italy
| | - Ione Tamagnini
- Pathology Unit, Azienda USL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Matteo Tudini
- Unit of Legal Medicine, Department of Medical and Surgical Sciences, University of Bologna, Via Irnerio 49, 40126 Bologna, Italy
| | - Paolo Fais
- Unit of Legal Medicine, Department of Medical and Surgical Sciences, University of Bologna, Via Irnerio 49, 40126 Bologna, Italy
| | - Susi Pelotti
- Unit of Legal Medicine, Department of Medical and Surgical Sciences, University of Bologna, Via Irnerio 49, 40126 Bologna, Italy
| |
Collapse
|
184
|
Wu Z, Cao Z, Yao H, Yan X, Xu W, Zhang M, Jiao Z, Zhang Z, Chen J, Liu Y, Zhang M, Wang D. Coupled deglycosylation-ubiquitination cascade in regulating PD-1 degradation by MDM2. Cell Rep 2023; 42:112693. [PMID: 37379210 DOI: 10.1016/j.celrep.2023.112693] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 05/02/2023] [Accepted: 06/09/2023] [Indexed: 06/30/2023] Open
Abstract
Posttranslational modifications represent a key step in modulating programmed death-1 (PD-1) functions, but the underlying mechanisms remain incompletely defined. Here, we report crosstalk between deglycosylation and ubiquitination in regulating PD-1 stability. We show that the removal of N-linked glycosylation is a prerequisite for efficient PD-1 ubiquitination and degradation. Murine double minute 2 (MDM2) is identified as an E3 ligase of deglycosylated PD-1. In addition, the presence of MDM2 facilitates glycosylated PD-1 interaction with glycosidase NGLY1 and promotes subsequent NGLY1-catalyzed PD-1 deglycosylation. Functionally, we demonstrate that the absence of T cell-specific MDM2 accelerates tumor growth by primarily upregulating PD-1. By stimulating the p53-MDM2 axis, interferon-α (IFN-α) reduces PD-1 levels in T cells, which, in turn, exhibit a synergistic effect on tumor suppression by sensitizing anti-PD-1 immunotherapy. Our study reveals that MDM2 directs PD-1 degradation via a deglycosylation-ubiquitination coupled mechanism and sheds light on a promising strategy to boost cancer immunotherapy by targeting the T cell-specific MDM2-PD-1 regulatory axis.
Collapse
Affiliation(s)
- Zhen Wu
- State Key Laboratory of Medical Molecular Biology & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Zhijie Cao
- State Key Laboratory of Medical Molecular Biology & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Han Yao
- State Key Laboratory of Medical Molecular Biology & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Xiaojun Yan
- State Key Laboratory of Medical Molecular Biology & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Wenbin Xu
- State Key Laboratory of Medical Molecular Biology & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Mi Zhang
- Department of Anatomy, Histology and Embryology, School of Basic Medicine, China Medical University, Shenyang 110122, China
| | - Zishan Jiao
- State Key Laboratory of Medical Molecular Biology & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Zijing Zhang
- State Key Laboratory of Medical Molecular Biology & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Jianyuan Chen
- State Key Laboratory of Medical Molecular Biology & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Yajing Liu
- State Key Laboratory of Medical Molecular Biology & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Meng Zhang
- State Key Laboratory of Medical Molecular Biology & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Donglai Wang
- State Key Laboratory of Medical Molecular Biology & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China.
| |
Collapse
|
185
|
Affiliation(s)
- Ashley Wysong
- From the Department of Dermatology, University of Nebraska Medical Center, Omaha
| |
Collapse
|
186
|
Hulen TM, Friese C, Kristensen NP, Granhøj JS, Borch TH, Peeters MJW, Donia M, Andersen MH, Hadrup SR, Svane IM, Met Ö. Ex vivo modulation of intact tumor fragments with anti-PD-1 and anti-CTLA-4 influences the expansion and specificity of tumor-infiltrating lymphocytes. Front Immunol 2023; 14:1180997. [PMID: 37359554 PMCID: PMC10285209 DOI: 10.3389/fimmu.2023.1180997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/23/2023] [Indexed: 06/28/2023] Open
Abstract
Checkpoint inhibition (CPI) therapy and adoptive cell therapy with autologous tumor-infiltrating lymphocytes (TIL-based ACT) are the two most effective immunotherapies for the treatment of metastatic melanoma. While CPI has been the dominating therapy in the past decade, TIL-based ACT is beneficial for individuals even after progression on previous immunotherapies. Given that notable differences in response have been made when used as a subsequent treatment, we investigated how the qualities of TILs changed when the ex vivo microenvironment of intact tumor fragments were modulated with checkpoint inhibitors targeting programmed death receptor 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4). Initially, we show that unmodified TILs from CPI-resistant individuals can be produced, are overwhelmingly terminally differentiated, and are capable of responding to tumor. We then investigate these properties in ex vivo checkpoint modulated TILs finding that that they retain these qualities. Lastly, we confirmed the specificity of the TILs to the highest responding tumor antigens, and identified this reactivity resides largely in CD39+CD69+ terminally differentiated populations. Overall, we found that anti-PD-1 will alter the proliferative capacity while anti-CTLA4 will influence breadth of antigen specificity.
Collapse
Affiliation(s)
- Thomas Morgan Hulen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Christina Friese
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | | | - Joachim Stoltenborg Granhøj
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Troels Holz Borch
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Marlies J. W. Peeters
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Marco Donia
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Mads Hald Andersen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Sine Reker Hadrup
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Özcan Met
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
187
|
Fang Y, Zhang X, Huang H, Zeng Z. The interplay between noncoding RNAs and drug resistance in hepatocellular carcinoma: the big impact of little things. J Transl Med 2023; 21:369. [PMID: 37286982 DOI: 10.1186/s12967-023-04238-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 05/30/2023] [Indexed: 06/09/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the leading cause of cancer-related death in people, and a common primary liver cancer. Lacking early diagnosis and a high recurrence rate after surgical resection, systemic treatment is still an important treatment method for advanced HCC. Different drugs have distinct curative effects, side effects and drug resistance due to different properties. At present, conventional molecular drugs for HCC have displayed some limitations, such as adverse drug reactions, insensitivity to some medicines, and drug resistance. Noncoding RNAs (ncRNAs), including microRNAs (miRNAs), long noncoding RNAs (lncRNAs) and circular RNAs (circRNAs), have been well documented to be involved in the occurrence and progression of cancer. Novel biomarkers and therapeutic targets, as well as research into the molecular basis of drug resistance, are urgently needed for the management of HCC. We review current research on ncRNAs and consolidate the known roles regulating drug resistance in HCC and examine the potential clinical applications of ncRNAs in overcoming drug resistance barriers in HCC based on targeted therapy, cell cycle non-specific chemotherapy and cell cycle specific chemotherapy.
Collapse
Affiliation(s)
- Yuan Fang
- Organ Transplantation Center, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming, 650032, Yunnan, People's Republic of China
| | - XiaoLi Zhang
- Gastrointestinal and Hernia Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - HanFei Huang
- Organ Transplantation Center, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming, 650032, Yunnan, People's Republic of China.
| | - Zhong Zeng
- Organ Transplantation Center, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming, 650032, Yunnan, People's Republic of China.
| |
Collapse
|
188
|
Dellino M, Cerbone M, Laganà AS, Vitagliano A, Vimercati A, Marinaccio M, Baldini GM, Malvasi A, Cicinelli E, Damiani GR, Cazzato G, Cascardi E. Upgrading Treatment and Molecular Diagnosis in Endometrial Cancer-Driving New Tools for Endometrial Preservation? Int J Mol Sci 2023; 24:9780. [PMID: 37298731 PMCID: PMC10253366 DOI: 10.3390/ijms24119780] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 06/12/2023] Open
Abstract
One emerging problem for onco-gynecologists is the incidence of premenopausal patients under 40 years of age diagnosed with stage I Endometrial Cancer (EC) who want to preserve their fertility. Our review aims to define a primary risk assessment that can help fertility experts and onco-gynecologists tailor personalized treatment and fertility-preserving strategies for fertile patients wishing to have children. We confirm that risk factors such as myometrial invasion and The International Federation of Gynecology and Obstetrics (FIGO) staging should be integrated into the novel molecular classification provided by The Cancer Genome Atlas (TCGA). We also corroborate the influence of classical risk factors such as obesity, Polycystic ovarian syndrome (PCOS), and diabetes mellitus to assess fertility outcomes. The fertility preservation options are inadequately discussed with women with a diagnosis of gynecological cancer. A multidisciplinary team of gynecologists, oncologists, and fertility specialists could increase patient satisfaction and improve fertility outcomes. The incidence and death rates of endometrial cancer are rising globally. International guidelines recommend radical hysterectomy and bilateral salpingo-oophorectomy as the standard of care for this cancer; however, fertility-sparing alternatives should be tailored to motivated women of reproductive age, establishing an appropriate cost-benefit balance between childbearing desire and cancer risk. New molecular classifications such as that of TCGA provide a robust supplementary risk assessment tool that can tailor the treatment options to the patient's needs, curtail over- and under-treatment, and contribute to the spread of fertility-preserving strategies.
Collapse
Affiliation(s)
- Miriam Dellino
- Obstetrics and Gynaecology Unit, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Marco Cerbone
- Obstetrics and Gynaecology Unit, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Antonio Simone Laganà
- Unit of Gynecologic Oncology, ARNAS “Civico—Di Cristina—Benfratelli”, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Amerigo Vitagliano
- Obstetrics and Gynaecology Unit, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Antonella Vimercati
- Obstetrics and Gynaecology Unit, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Marco Marinaccio
- Obstetrics and Gynaecology Unit, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | | | - Antonio Malvasi
- Obstetrics and Gynaecology Unit, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Ettore Cicinelli
- Obstetrics and Gynaecology Unit, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Gianluca Raffaello Damiani
- Obstetrics and Gynaecology Unit, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Gerardo Cazzato
- Department of Emergency and Organ Transplantation, Pathology Section, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Eliano Cascardi
- Department of Medical Sciences, University of Turin, 10124 Turin, Italy
- Pathology Unit, FPO-IRCCS Candiolo Cancer Institute, 10060 Candiolo, Italy
| |
Collapse
|
189
|
Song R, Liu F, Ping Y, Zhang Y, Wang L. Potential non-invasive biomarkers in tumor immune checkpoint inhibitor therapy: response and prognosis prediction. Biomark Res 2023; 11:57. [PMID: 37268978 DOI: 10.1186/s40364-023-00498-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 05/07/2023] [Indexed: 06/04/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have dramatically enhanced the treatment outcomes for diverse malignancies. Yet, only 15-60% of patients respond significantly. Therefore, accurate responder identification and timely ICI administration are critical issues in tumor ICI therapy. Recent rapid developments at the intersection of oncology, immunology, biology, and computer science have provided an abundance of predictive biomarkers for ICI efficacy. These biomarkers can be invasive or non-invasive, depending on the specific sample collection method. Compared with invasive markers, a host of non-invasive markers have been confirmed to have superior availability and accuracy in ICI efficacy prediction. Considering the outstanding advantages of dynamic monitoring of the immunotherapy response and the potential for widespread clinical application, we review the recent research in this field with the aim of contributing to the identification of patients who may derive the greatest benefit from ICI therapy.
Collapse
Affiliation(s)
- Ruixia Song
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou University, Zhengzhou, Henan, China
| | - Fengsen Liu
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou University, Zhengzhou, Henan, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Yu Ping
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yi Zhang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou University, Zhengzhou, Henan, China.
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, Henan, China.
| | - Liping Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
190
|
Najafi A, Sabaghi M, Asgarian-Omran H, Valadan R, Shekarriz R, Zaboli E, Janbabai G, Tehrani M. Relative Expression of BATF and CD112 in PBMC of Patients with Chronic Lymphocytic Leukemia. Asian Pac J Cancer Prev 2023; 24:2171-2176. [PMID: 37378949 PMCID: PMC10505859 DOI: 10.31557/apjcp.2023.24.6.2171] [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: 01/14/2023] [Accepted: 06/22/2023] [Indexed: 06/29/2023] Open
Abstract
OBJECTIVE BATF, as a transcription factor, and CD112, as a receptor for TIGIT, are involved in T-cell exhaustion. We investigated BATF and CD112 gene expression in the peripheral blood mononuclear cells from CLL patients and healthy subjects. METHODS In a case-control study, 33 patients with CLL and 20 sex- and age-matched healthy individual were enrolled. Diagnosis and classification of patients was done according to immunophenotyping via flow cytometry and RAI staging system, respectively. Relative mRNA expression of BATF and CD112 was measured using qRT-PCR. RESULT Our results showed that the expression of BATF and CD112 in CLL samples were significantly decreased in comparison those of the healthy controls (P = 0.0236 and P = 0.0002, respectively). CONCLUSION These findings suggest the role of BATF and CD112 not only as a role in T cell exhaustion, but in effector differentiation program in CLL, which warrants further studies in future.
Collapse
Affiliation(s)
- Ahmad Najafi
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Matineh Sabaghi
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Hossein Asgarian-Omran
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Reza Valadan
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Ramin Shekarriz
- Department of Hematology and Oncology, Imam Khomeini Hospital, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Ehsan Zaboli
- Department of Hematology and Oncology, Imam Khomeini Hospital, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Ghasem Janbabai
- Department of Hematology and Oncology, Imam Khomeini Hospital, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Mohsen Tehrani
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
191
|
Hamid O, Hassel JC, Shoushtari AN, Meier F, Bauer TM, Salama AKS, Kirkwood JM, Ascierto PA, Lorigan PC, Mauch C, Orloff M, Evans TRJ, Holland C, Edukulla R, Abedin SE, Middleton MR. Tebentafusp in combination with durvalumab and/or tremelimumab in patients with metastatic cutaneous melanoma: a phase 1 study. J Immunother Cancer 2023; 11:e006747. [PMID: 37286303 PMCID: PMC10254987 DOI: 10.1136/jitc-2023-006747] [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] [Accepted: 05/12/2023] [Indexed: 06/09/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors have significantly improved outcomes in first line cutaneous melanoma. However, there is a high unmet need for patients who progress on these therapies and combination therapies are being explored to improve outcomes. Tebentafusp is a first-in-class gp100×CD3 ImmTAC bispecific that demonstrated overall survival (OS) benefit (HR 0.51) in metastatic uveal melanoma despite a modest overall response rate of 9%. This phase 1b trial evaluated the safety and initial efficacy of tebentafusp in combination with durvalumab (anti-programmed death ligand 1 (PDL1)) and/or tremelimumab (anti-cytotoxic T lymphocyte-associated antigen 4) in patients with metastatic cutaneous melanoma (mCM), the majority of whom progressed on prior checkpoint inhibitors. METHODS In this open-label, multicenter, phase 1b, dose-escalation trial, HLA-A*02:01-positive patients with mCM received weekly intravenous tebentafusp with increasing monthly doses of durvalumab and/or tremelimumab starting day 15 of each cycle. The primary objective was to identify the maximum tolerated dose (MTD) or recommended phase 2 dose for each combination. Efficacy analyses were performed in all tebentafusp with durvalumab±tremelimumab treated patients with a sensitivity analysis in those who progressed on prior anti-PD(L)1 therapy. RESULTS 85 patients were assigned to receive tebentafusp in combination with durvalumab (n=43), tremelimumab (n=13), or durvalumab and tremelimumab (n=29). Patients were heavily pretreated with a median of 3 prior lines of therapy, including 76 (89%) who received prior anti-PD(L)1. Maximum target doses of tebentafusp (68 mcg) alone or in combination with durvalumab (20 mg/kg) and tremelimumab (1 mg/kg) were tolerated; MTD was not formally identified for any arm. Adverse event profile was consistent with each individual therapy and there were no new safety signals nor treatment-related deaths. In the efficacy subset (n=72), the response rate was 14%, tumor shrinkage rate was 41% and 1-year OS rate was 76% (95% CI: 70% to 81%). The 1-year OS for triplet combination (79%; 95% CI: 71% to 86%) was similar to tebentafusp plus durvalumab (74%; 95% CI: 67% to 80%). CONCLUSION At maximum target doses, the safety of tebentafusp with checkpoint inhibitors was consistent with safety of each individual therapy. Tebentafusp with durvalumab demonstrated promising efficacy in heavily pretreated patients with mCM, including those who progressed on prior anti-PD(L)1. TRIAL REGISTRATION NUMBER NCT02535078.
Collapse
Affiliation(s)
- Omid Hamid
- The Angeles Clinic and Research Institute, a Cedars-Sinai Affiliate, Los Angeles, California, USA
| | | | - Alexander N Shoushtari
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Weill Cornell Medical College, New York, New York, USA
| | - Friedegund Meier
- Skin Cancer Center at the National Center for Tumor Diseases and University Cancer Centre, Dresden, Germany
- Department of Dermatology, University Hospital Carl Gustav Carus, Dresden, Germany
| | | | | | - John M Kirkwood
- University of Pittsburgh Medical Center Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | | | | | - Marlana Orloff
- Sidney Kimmel Cancer Center, Jefferson University, Philadelphia, Pennsylvania, USA
| | | | | | | | | | - Mark R Middleton
- Department of Oncology, Medical Sciences Division, University of Oxford, Oxford, UK
| |
Collapse
|
192
|
Disis ML, Adams SF, Bajpai J, Butler MO, Curiel T, Dodt SA, Doherty L, Emens LA, Friedman CF, Gatti-Mays M, Geller MA, Jazaeri A, John VS, Kurnit KC, Liao JB, Mahdi H, Mills A, Zsiros E, Odunsi K. Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immunotherapy for the treatment of gynecologic cancer. J Immunother Cancer 2023; 11:e006624. [PMID: 37295818 PMCID: PMC10277149 DOI: 10.1136/jitc-2022-006624] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2023] [Indexed: 06/12/2023] Open
Abstract
Advanced gynecologic cancers have historically lacked effective treatment options. Recently, immune checkpoint inhibitors (ICIs) have been approved by the US Food and Drug Administration for the treatment of cervical cancer and endometrial cancer, offering durable responses for some patients. In addition, many immunotherapy strategies are under investigation for the treatment of earlier stages of disease or in other gynecologic cancers, such as ovarian cancer and rare gynecologic tumors. While the integration of ICIs into the standard of care has improved outcomes for patients, their use requires a nuanced understanding of biomarker testing, treatment selection, patient selection, response evaluation and surveillance, and patient quality of life considerations, among other topics. To address this need for guidance, the Society for Immunotherapy of Cancer (SITC) convened a multidisciplinary panel of experts to develop a clinical practice guideline. The Expert Panel drew on the published literature as well as their own clinical experience to develop evidence- and consensus-based recommendations to provide guidance to cancer care professionals treating patients with gynecologic cancer.
Collapse
Affiliation(s)
- Mary L Disis
- Cancer Vaccine Institute, University of Washington, Seattle, Washington, USA
| | - Sarah F Adams
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, The University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico, USA
| | - Jyoti Bajpai
- Medical Oncology, Tata Memorial Centre, Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Marcus O Butler
- Department of Medical Oncology and Hematology, Princess Margaret Hospital Cancer Centre, Toronto, Ontario, Canada
| | - Tyler Curiel
- Dartmouth-Hitchcock's Norris Cotton Cancer Center, Dartmouth Medical School, Hanover, New Hampshire, USA
| | | | - Laura Doherty
- Program in Women's Oncology, Women and Infants Hospital of Rhode Island, Providence, Rhode Island, USA
| | - Leisha A Emens
- Department of Medicine, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Claire F Friedman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Margaret Gatti-Mays
- Pelotonia Institute for Immuno-Oncology, Division of Medical Oncology, The Ohio State University, Columbus, Ohio, USA
| | - Melissa A Geller
- Department of Obstetrics, Gynecology & Women's Health, Division of Gynecologic Oncology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Amir Jazaeri
- Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Veena S John
- Department of Medical Oncology & Hematology, Northwell Health Cancer Institute, Lake Success, New York, USA
| | - Katherine C Kurnit
- University of Chicago Medicine Comprehensive Cancer Center, University of Chicago, Chicago, Illinois, USA
| | - John B Liao
- University of Washington School of Medicine, Seattle, Washington, USA
| | - Haider Mahdi
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Anne Mills
- Department of Pathology, University of Virginia Health System, Charlottesville, Virginia, USA
| | - Emese Zsiros
- Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Kunle Odunsi
- The University of Chicago Medicine Comprehensive Cancer Center, Chicago, Illinois, USA
| |
Collapse
|
193
|
Xu S, Wang C, Yang L, Wu J, Li M, Xiao P, Xu Z, Xu Y, Wang K. Targeting immune checkpoints on tumor-associated macrophages in tumor immunotherapy. Front Immunol 2023; 14:1199631. [PMID: 37313405 PMCID: PMC10258331 DOI: 10.3389/fimmu.2023.1199631] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/16/2023] [Indexed: 06/15/2023] Open
Abstract
Unprecedented breakthroughs have been made in cancer immunotherapy in recent years. Particularly immune checkpoint inhibitors have fostered hope for patients with cancer. However, immunotherapy still exhibits certain limitations, such as a low response rate, limited efficacy in certain populations, and adverse events in certain tumors. Therefore, exploring strategies that can improve clinical response rates in patients is crucial. Tumor-associated macrophages (TAMs) are the predominant immune cells that infiltrate the tumor microenvironment and express a variety of immune checkpoints that impact immune functions. Mounting evidence indicates that immune checkpoints in TAMs are closely associated with the prognosis of patients with tumors receiving immunotherapy. This review centers on the regulatory mechanisms governing immune checkpoint expression in macrophages and strategies aimed at improving immune checkpoint therapies. Our review provides insights into potential therapeutic targets to improve the efficacy of immune checkpoint blockade and key clues to developing novel tumor immunotherapies.
Collapse
Affiliation(s)
- Shumin Xu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Chenyang Wang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Lingge Yang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Jiaji Wu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Mengshu Li
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Peng Xiao
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhiyong Xu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Yun Xu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Kai Wang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| |
Collapse
|
194
|
Mannucci M, Fontana V, Campanella D, Filiberti RA, Pronzato P, Rosa A. A Descriptive Study of Repeated Hospitalizations and Survival of Patients with Metastatic Melanoma in the Northern Italian Region during 2004-2019. Curr Oncol 2023; 30:5266-5278. [PMID: 37366883 DOI: 10.3390/curroncol30060400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/17/2023] [Accepted: 05/23/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND Survival rates for metastatic melanoma (MM) patients have improved in recent years, leading to major expenses and health resource use. We conducted a non-concurrent prospective study to describe the burden of hospitalization in a real-world setting for patients with MM. METHODS Patients were tracked throughout all hospital stays in 2004-2019 by means of hospital discharges. The number of hospitalizations, the rehospitalization rate, the average time spent in the hospital and the time span between consecutive admissions were evaluated. Relative survival was also calculated. RESULTS Overall, 1570 patients were identified at the first stay (56.5% in 2004-2011 and 43.7% in 2012-2019). A total of 8583 admissions were retrieved. The overall rehospitalization rate was 1.78 per patient/year (95%CI = 1.68-1.89); it increased significantly with the period of first stay (1.51, 95%CI = 1.40-1.64 in 2004-2011 and 2.11, 95%CI = 1.94-2.29 thereafter). The median time span between hospitalizations was lower for patients hospitalized after 2011 (16 vs. 26 months). An improvement in survival for males was highlighted. CONCLUSIONS The hospitalization rate of patients with MM was higher in the last years of the study. Compared with a shorter length of stay, patients were admitted to hospitals with a higher frequency. Knowledge of the burden of MM is essential for planning the allocation of healthcare resources.
Collapse
Affiliation(s)
- Matilde Mannucci
- Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Vincenzo Fontana
- Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Dalila Campanella
- Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Rosa Angela Filiberti
- Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Paolo Pronzato
- Medical Oncology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Alessandra Rosa
- Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| |
Collapse
|
195
|
Chen J, Wang D, Chan S, Yang Q, Wang C, Wang X, Sun R, Gui Y, Yu S, Yang J, Zhang H, Zhang X, Tang K, Zhang H, Liu S. Development and validation of a novel T cell proliferation-related prognostic model for predicting survival and immunotherapy benefits in melanoma. Aging (Albany NY) 2023; 15:204748. [PMID: 37227816 DOI: 10.18632/aging.204748] [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: 02/28/2023] [Accepted: 05/09/2023] [Indexed: 05/27/2023]
Abstract
BACKGROUND T cell plays a crucial role in the occurrence and progression of Skin cutaneous melanoma (SKCM). This research aims to identify the actions of T cell proliferation-related genes (TRGs) on the prognosis and immunotherapy response of tumor patients. METHOD The clinical manifestation and gene expression data of SKCM patients were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. T cell proliferation-related molecular subtypes were identified utilizing consensus clustering. Subsequently, Cox and Lasso regression analysis was conducted to identify six prognostic genes, and a prognostic signature was constructed. A series of experiments, such as qRT-PCR, Western blotting and CCK8 assay, were then conducted to verify the reliability of the six genes. RESULTS In this study, a grading system was established to forecast survival time and responses to immunotherapy, providing an overview of the tumoral immune landscape. Meanwhile, we identified six prognostic signature genes. Notably, we also found that C1RL protein may inhibit the growth of melanoma cell lines. CONCLUSION The scoring system depending on six prognostic genes showed great efficiency in predicting survival time. The system could help to forecast prognosis of SKCM patients, characterize SKCM immunological condition, assess patient immunotherapy response.
Collapse
Affiliation(s)
- Jiajie Chen
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230022, China
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui 230022, China
| | - Daiyue Wang
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230022, China
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui 230022, China
| | - Shixin Chan
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Qingqing Yang
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230022, China
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui 230022, China
| | - Chen Wang
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230022, China
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui 230022, China
| | - Xu Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Rui Sun
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Yu Gui
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230022, China
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui 230022, China
| | - Shuling Yu
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230022, China
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui 230022, China
| | - Jinwei Yang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Haoxue Zhang
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230022, China
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui 230022, China
| | - Xiaomin Zhang
- Department of Biochemistry and Molecular Biology, Metabolic Disease Research Center, School of Basic Medicine, Anhui Medical University, Hefei, Anhui 230022, China
| | - Kechao Tang
- Department of Biochemistry and Molecular Biology, Metabolic Disease Research Center, School of Basic Medicine, Anhui Medical University, Hefei, Anhui 230022, China
| | - Huabing Zhang
- Affiliated Chuzhou Hospital of Anhui Medical University, The First People’s Hospital of Chuzhou, Chuzhou, Anhui 230022, China
- Department of Biochemistry and Molecular Biology, Metabolic Disease Research Center, School of Basic Medicine, Anhui Medical University, Hefei, Anhui 230022, China
| | - Shengxiu Liu
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui 230022, China
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui 230022, China
| |
Collapse
|
196
|
Du F, Yang LH, Liu J, Wang J, Fan L, Duangmano S, Liu H, Liu M, Wang J, Zhong X, Zhang Z, Wang F. The role of mitochondria in the resistance of melanoma to PD-1 inhibitors. J Transl Med 2023; 21:345. [PMID: 37221594 DOI: 10.1186/s12967-023-04200-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 05/14/2023] [Indexed: 05/25/2023] Open
Abstract
Malignant melanoma is one of the most common tumours and has the highest mortality rate of all types of skin cancers worldwide. Traditional and novel therapeutic approaches, including surgery, targeted therapy and immunotherapy, have shown good efficacy in the treatment of melanoma. At present, the mainstay of treatment for melanoma is immunotherapy combined with other treatment strategies. However, immune checkpoint inhibitors, such as PD-1 inhibitors, are not particularly effective in the clinical treatment of patients with melanoma. Changes in mitochondrial function may affect the development of melanoma and the efficacy of PD-1 inhibitors. To elucidate the role of mitochondria in the resistance of melanoma to PD-1 inhibitors, this review comprehensively summarises the role of mitochondria in the occurrence and development of melanoma, targets related to the function of mitochondria in melanoma cells and changes in mitochondrial function in different cells in melanoma resistant to PD-1 inhibitors. This review may help to develop therapeutic strategies for improving the clinical response rate of PD-1 inhibitors and prolonging the survival of patients by activating mitochondrial function in tumour and T cells.
Collapse
Affiliation(s)
- Fei Du
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Lu-Han Yang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Jiao Liu
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
- Department of Pharmacy, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Jian Wang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Lianpeng Fan
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Suwit Duangmano
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Hao Liu
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Minghua Liu
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Jun Wang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Xiaolin Zhong
- Department of Pharmacy, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Zhuo Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China.
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand.
| | - Fang Wang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China.
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
197
|
Stoff R, Asher N, Laks S, Steinberg Y, Schachter J, Shapira-Frommer R, Grynberg S, Ben-Betzalel G. Real world evidence of Lenvatinib + anti PD-1 as an advanced line for metastatic melanoma. Front Oncol 2023; 13:1180988. [PMID: 37274272 PMCID: PMC10233023 DOI: 10.3389/fonc.2023.1180988] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 05/08/2023] [Indexed: 06/06/2023] Open
Abstract
Introduction Immunotherapy has revolutionized the prognosis of patients with metastatic melanoma. To date, the most active regimen is the combination of ipilimumab + nivolumab (ipi-nivo) achieving a response rate of nearly 60% and a median survival (OS) of 6 years. However, approximately 40% of patients experience primary resistance, while around 50% experience secondary resistance, highlighting the need for an effective second-line treatment option The recently published results on the use of lenvatinib + pembrolizumab in the advanced line setting led to the adoption of this regimen at our institution. Here we present our experience with this regimen, focusing on efficacy and safety. Methods Electronic medical records of patients treated at a tertiary referral melanoma center, with at least one cycle of anti PD-1 + lenvatinib from 2020 to 2023 were analyzed for baseline demographic characteristics, disease related characteristics and treatment outcomes. Results Forty-two patients were identified. The Response rate (RR) was 28% and the disease control rate was 38%. Responses were seen across different melanoma subtypes, including 67% in acral melanoma, 20% in uveal melanoma, and 25% in mucosal melanoma. Patients with a more aggressive disease manifested by elevated lactate dehydrogenase (LDH) achieved a RR of 26%, while patients with active central nervous system (CNS) metastases had a RR of 31%, and an intra-cranial RR of 23%. Responses were seen across lines of treatment, with a 25% RR in the second and third lines, and a 36% RR in the fourth and fifth lines. The median progression free survival was 3 months, and the median survival was 11 months. The treatment was not easily tolerated with 31% of the patients experiencing grade 3-4 toxicity, which was manageable through dose interruptions and reductions. Only 7% of patients discontinued the treatment due to toxicity. Conclusion Lenvatinib in combination with anti-PD1 had demonstrated both relative safety and efficacy in patients with metastatic melanoma of all subtypes in the advanced line setting. We are eagerly anticipating the mature results of the LEAP-004 study hoping that this regimen will receive regulatory approval, paving the way for its widespread adoption in daily practice worldwide.
Collapse
|
198
|
Dulal D, Boring A, Terrero D, Johnson T, Tiwari AK, Raman D. Tackling of Immunorefractory Tumors by Targeting Alternative Immune Checkpoints. Cancers (Basel) 2023; 15:2774. [PMID: 37345111 PMCID: PMC10216651 DOI: 10.3390/cancers15102774] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/09/2023] [Accepted: 05/12/2023] [Indexed: 06/23/2023] Open
Abstract
Physiologically, well known or traditional immune checkpoints (ICs), such as CTLA-4 and PD-1, are in place to promote tolerance to self-antigens and prevent generation of autoimmunity. In cancer, the ICs are effectively engaged by the tumor cells or stromal ells from the tumor microenvironment through expression of cognate ligands for the ICs present on the cell surface of CD8+ T lymphocytes. The ligation of ICs on CD8+ T lymphocytes triggers inhibitory signaling pathways, leading to quiescence or an exhaustion of CD8+ T lymphocytes. This results in failure of immunotherapy. To overcome this, several FDA-approved therapeutic antibodies are available, but the clinical outcome is quite variable due to the resistance encountered through upregulated expression of alternate ICs such as VISTA, LAG-3, TIGIT and TIM-3. This review focuses on the roles played by the traditional as well as alternate ICs and the contribution of associated signaling pathways in generating such resistance to immunotherapy. Combinatorial targeting of traditional and alternate ICs might be beneficial for immune-refractory tumors.
Collapse
Affiliation(s)
- Dharmindra Dulal
- Department of Cell and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH 43614, USA; (D.D.); (A.B.); (A.K.T.)
| | - Andrew Boring
- Department of Cell and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH 43614, USA; (D.D.); (A.B.); (A.K.T.)
| | - David Terrero
- Department of Pharmacology & Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo Main Campus, Toledo, OH 43614, USA
| | - Tiffany Johnson
- Department of Cell and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH 43614, USA; (D.D.); (A.B.); (A.K.T.)
| | - Amit K. Tiwari
- Department of Cell and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH 43614, USA; (D.D.); (A.B.); (A.K.T.)
- Department of Pharmacology & Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo Main Campus, Toledo, OH 43614, USA
| | - Dayanidhi Raman
- Department of Cell and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH 43614, USA; (D.D.); (A.B.); (A.K.T.)
| |
Collapse
|
199
|
Ziogas DC, Theocharopoulos C, Lialios PP, Foteinou D, Koumprentziotis IA, Xynos G, Gogas H. Beyond CTLA-4 and PD-1 Inhibition: Novel Immune Checkpoint Molecules for Melanoma Treatment. Cancers (Basel) 2023; 15:2718. [PMID: 37345056 PMCID: PMC10216291 DOI: 10.3390/cancers15102718] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/04/2023] [Accepted: 05/08/2023] [Indexed: 06/23/2023] Open
Abstract
More than ten years after the approval of ipilimumab, immune checkpoint inhibitors (ICIs) against PD-1 and CTLA-4 have been established as the most effective treatment for locally advanced or metastatic melanoma, achieving durable responses either as monotherapies or in combinatorial regimens. However, a considerable proportion of patients do not respond or experience early relapse, due to multiple parameters that contribute to melanoma resistance. The expression of other immune checkpoints beyond the PD-1 and CTLA-4 molecules remains a major mechanism of immune evasion. The recent approval of anti-LAG-3 ICI, relatlimab, in combination with nivolumab for metastatic disease, has capitalized on the extensive research in the field and has highlighted the potential for further improvement of melanoma prognosis by synergistically blocking additional immune targets with new ICI-doublets, antibody-drug conjugates, or other novel modalities. Herein, we provide a comprehensive overview of presently published immune checkpoint molecules, including LAG-3, TIGIT, TIM-3, VISTA, IDO1/IDO2/TDO, CD27/CD70, CD39/73, HVEM/BTLA/CD160 and B7-H3. Beginning from their immunomodulatory properties as co-inhibitory or co-stimulatory receptors, we present all therapeutic modalities targeting these molecules that have been tested in melanoma treatment either in preclinical or clinical settings. Better understanding of the checkpoint-mediated crosstalk between melanoma and immune effector cells is essential for generating more effective strategies with augmented immune response.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Helen Gogas
- First Department of Medicine, Laiko General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.T.); (P.-P.L.); (D.F.); (I.-A.K.); (G.X.)
| |
Collapse
|
200
|
Galati D, Zanotta S, Capone M, Madonna G, Mallardo D, Romanelli M, Simeone E, Festino L, Sparano F, Azzaro R, De Filippi R, Pinto A, Paulos CM, Ascierto PA. Potential clinical implications of CD4 +CD26 high T cells for nivolumab treated melanoma patients. J Transl Med 2023; 21:318. [PMID: 37170241 PMCID: PMC10176780 DOI: 10.1186/s12967-023-04184-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 05/05/2023] [Indexed: 05/13/2023] Open
Abstract
BACKGROUND Nivolumab is an anti-PD1 antibody that has dramatically improved metastatic melanoma patients' outcomes. Nevertheless, many patients are resistant to PD-1 inhibition, occasionally experiencing severe off-target immune toxicity. In addition, no robust and reproducible biomarkers have yet been validated to identify the correct selection of patients who will benefit from anti-PD-1 treatment avoiding unwanted side effects. However, the strength of CD26 expression on CD4+ T lymphocytes permits the characterization of three subtypes with variable degrees of responsiveness to tumors, suggesting that the presence of CD26-expressing T cells in patients might be a marker of responsiveness to PD-1-based therapies. METHODS The frequency distribution of peripheral blood CD26-expressing cells was investigated employing multi-parametric flow cytometry in 69 metastatic melanoma patients along with clinical characteristics and blood count parameters at baseline (W0) and compared to 20 age- and sex-matched healthy controls. Percentages of baseline CD4+CD26high T cells were correlated with the outcome after nivolumab treatment. In addition, the frequency of CD4+CD26high T cells at W0 was compared with those obtained after 12 weeks (W1) of therapy in a sub-cohort of 33 patients. RESULTS Circulating CD4+CD26high T cells were significantly reduced in melanoma patients compared to healthy subjects (p = 0.001). In addition, a significant association was observed between a low baseline percentage of CD4+CD26high T cells (< 7.3%) and clinical outcomes, measured as overall survival (p = 0.010) and progression-free survival (p = 0.014). Moreover, patients with clinical benefit from nivolumab therapy had significantly higher frequencies of circulating CD4+CD26high T cells than patients with non-clinical benefit (p = 0.004) at 12 months. Also, a higher pre-treatment proportion of circulating CD4+CD26high T cells was correlated with Disease Control Rate (p = 0.014) and best Overall Response Rate (p = 0.009) at 12 months. Interestingly, after 12 weeks (W1) of nivolumab treatment, percentages of CD4+CD26high T cells were significantly higher in comparison with the frequencies measured at W0 (p < 0.0001), aligning the cell counts with the ranges seen in the blood of healthy subjects. CONCLUSIONS Our study firstly demonstrates that peripheral blood circulating CD4+CD26high T lymphocytes represent potential biomarkers whose perturbations are associated with reduced survival and worse clinical outcomes in melanoma patients.
Collapse
Affiliation(s)
- Domenico Galati
- Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
- Hematology-Oncology and Stem Cell Transplantation Unit, Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
| | - Serena Zanotta
- Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
- Hematology-Oncology and Stem Cell Transplantation Unit, Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
| | - Mariaelena Capone
- Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
| | - Gabriele Madonna
- Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
| | - Domenico Mallardo
- Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
| | - Marilena Romanelli
- Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
| | - Ester Simeone
- Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
| | - Lucia Festino
- Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
| | - Francesca Sparano
- Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
| | - Rosa Azzaro
- Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
| | - Rosaria De Filippi
- Dipartimento di Medicina Clinica e Chirurgia, Università Degli Studi Federico II, Naples, Italy
| | - Antonio Pinto
- Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
- Hematology-Oncology and Stem Cell Transplantation Unit, Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
| | - Chrystal M. Paulos
- Division of Surgical Oncology, Department of Surgery, Emory University, Atlanta, GA USA
- Department of Microbiology and Immunology, Winship Cancer Institute, Emory University, Atlanta, GA USA
| | - Paolo A. Ascierto
- Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, Naples, Italy
| |
Collapse
|