751
|
Mandalà M, Rutkowski P. Rational combination of cancer immunotherapy in melanoma. Virchows Arch 2018; 474:433-447. [PMID: 30552520 DOI: 10.1007/s00428-018-2506-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 12/04/2018] [Indexed: 12/15/2022]
Abstract
The recent advances in cancer immunotherapy with unprecedented success in therapy of advanced melanoma represent a turning point in the landscape of melanoma treatment. Given the complexity of activation of immunological system and the physiologic multifactorial homeostatic mechanisms controlling immune responses, combinatorial strategies are eagerly needed in melanoma therapy. Nevertheless, rational selection of immunotherapy combinations should be more biomarker-guided, including not only the cancer immunogram, PD-L1 expression, interferon gene expression signature, mutational burden, and tumor infiltration by CD8+ T cells but also intratumoral T cell exhaustion and microbiota composition. In this review, we summarize the rationale to develop combination treatment strategies in melanoma and discuss biological background that could help to design new combinations in order to improve patients' outcome.
Collapse
Affiliation(s)
- Mario Mandalà
- Unit of Medical Oncology, Department of Oncology and Haematology, Papa Giovanni XXIII Cancer Center Hospital, Piazza OMS 1, 24100, Bergamo, Italy.
| | - Piotr Rutkowski
- Maria Sklodowska-Curie Institute, Oncology Center, Warsaw, Poland
| |
Collapse
|
752
|
Racz JM, Block MS, Baum CL, Jakub JW. Management of local or regional non‐nodal disease. J Surg Oncol 2018; 119:187-199. [DOI: 10.1002/jso.25330] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 11/19/2018] [Indexed: 12/31/2022]
|
753
|
Hauschild A, Dummer R, Schadendorf D, Santinami M, Atkinson V, Mandalà M, Chiarion-Sileni V, Larkin J, Nyakas M, Dutriaux C, Haydon A, Robert C, Mortier L, Schachter J, Lesimple T, Plummer R, Dasgupta K, Haas T, Shilkrut M, Gasal E, Kefford R, Kirkwood JM, Long GV. Longer Follow-Up Confirms Relapse-Free Survival Benefit With Adjuvant Dabrafenib Plus Trametinib in Patients With Resected BRAF V600-Mutant Stage III Melanoma. J Clin Oncol 2018; 36:3441-3449. [PMID: 30343620 PMCID: PMC6286159 DOI: 10.1200/jco.18.01219] [Citation(s) in RCA: 180] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
PURPOSE Dabrafenib plus trametinib improved relapse-free survival (RFS) versus placebo (hazard ratio [HR], 0.47; P < .001) in patients with resected BRAF V600-mutant stage III melanoma (BRF115532; COMBI-AD; ClinicalTrials.gov identifier: NCT01682083). We present an updated RFS analysis on the basis of extended study follow-up and a cure-rate model analysis to estimate the fraction of patients expected to remain relapse free long term. METHODS In this phase III trial, patients with resected BRAF V600-mutant stage III melanoma were randomly assigned to 12 months of adjuvant dabrafenib plus trametinib versus placebo. We report updated RFS (primary end point) and distant metastasis-free survival. RFS was also analyzed by subgroups defined by baseline disease stage (American Joint Committee on Cancer 7th and 8th editions), nodal metastatic burden, and ulceration status. The fraction of patients who remained relapse free long term was estimated using a Weibull mixture cure-rate model. RESULTS At median follow-up of 44 months (dabrafenib plus trametinib) and 42 months (placebo), 3- and 4-year RFS rates were 59% (95% CI, 55% to 64%) and 54% (95% CI, 49% to 59%) in the dabrafenib plus trametinib arm and 40% (95% CI, 35% to 45%) and 38% (95% CI, 34% to 44%) in the placebo arm, respectively (HR, 0.49; 95% CI, 0.40 to 0.59). Distant metastasis-free survival also favored dabrafenib plus trametinib (HR, 0.53; 95% CI, 0.42 to 0.67). The estimated cure rate was 54% (95% CI, 49% to 59%) in the dabrafenib plus trametinib arm compared with 37% (95% CI, 32% to 42%) in the placebo arm. Subgroup analysis of RFS demonstrated similar treatment benefit regardless of baseline factors, including disease stage, nodal metastatic burden, and ulceration. CONCLUSION Longer follow-up confirmed RFS benefit with dabrafenib plus trametinib. Subgroup analysis suggested that dabrafenib plus trametinib benefited patients regardless of baseline factors.
Collapse
Affiliation(s)
- Axel Hauschild
- Axel Hauschild, University Hospital Schleswig-Holstein, Kiel; Dirk Schadendorf, University Hospital Essen, Essen; German Cancer Consortium, Heidelberg, Germany; Reinhard Dummer, University Hospital Zürich Skin Cancer Center, Zürich; Tomas Haas, Novartis AG, Basel, Switzerland; Mario Santinami, Fondazione Istituto Nazionale Tumori, Milan; Mario Mandalà, Papa Giovanni XXIII Cancer Center Hospital, Bergamo; Vanna Chiarion-Sileni, Veneto Institute of Oncology–Istituto di Ricovero e Cura a Carattere Scientifico, Padova, Italy; Victoria Atkinson, Princess Alexandra Hospital; Gallipoli Medical Research Foundation; University of Queensland, Brisbane, Queensland; Andrew Haydon, The Alfred Hospital, Melbourne, Victoria; Richard Kefford, Macquarie University; Westmead Hospital; Richard Kefford and Georgina V. Long, Melanoma Institute Australia; University of Sydney; Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia; James Larkin, Royal Marsden National Health Service Foundation Trust, London; Ruth Plummer, Freeman Hospital and Newcastle University, Newcastle upon Tyne, United Kingdom; Marta Nyakas, Oslo University Hospital, Oslo, Norway; Caroline Dutriaux, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint-André, Bordeaux; Caroline Robert, Institute Gustave Roussy, Paris; Laurent Mortier, Université de Lille, Institut National de la Santé et de la Recherche Médicale U1189, Centre Hospitalier Universitaire de sa Region Lille, Lille; Thierry Lesimple, Centre Eugène Marquis, Rennes, France; Jacob Schachter, Sheba Medical Center, Tel Hashomer; Tel Aviv University, Tel Aviv, Israel; Kohinoor Dasgupta, Novartis Healthcare, Hyderabad, India; Mark Shilkrut and Eduard Gasal, Novartis Pharmaceuticals Corporation, East Hanover, NJ; and John M. Kirkwood, UPMC Hillman Cancer Center; University of Pittsburgh, Pittsburgh, PA
| | - Reinhard Dummer
- Axel Hauschild, University Hospital Schleswig-Holstein, Kiel; Dirk Schadendorf, University Hospital Essen, Essen; German Cancer Consortium, Heidelberg, Germany; Reinhard Dummer, University Hospital Zürich Skin Cancer Center, Zürich; Tomas Haas, Novartis AG, Basel, Switzerland; Mario Santinami, Fondazione Istituto Nazionale Tumori, Milan; Mario Mandalà, Papa Giovanni XXIII Cancer Center Hospital, Bergamo; Vanna Chiarion-Sileni, Veneto Institute of Oncology–Istituto di Ricovero e Cura a Carattere Scientifico, Padova, Italy; Victoria Atkinson, Princess Alexandra Hospital; Gallipoli Medical Research Foundation; University of Queensland, Brisbane, Queensland; Andrew Haydon, The Alfred Hospital, Melbourne, Victoria; Richard Kefford, Macquarie University; Westmead Hospital; Richard Kefford and Georgina V. Long, Melanoma Institute Australia; University of Sydney; Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia; James Larkin, Royal Marsden National Health Service Foundation Trust, London; Ruth Plummer, Freeman Hospital and Newcastle University, Newcastle upon Tyne, United Kingdom; Marta Nyakas, Oslo University Hospital, Oslo, Norway; Caroline Dutriaux, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint-André, Bordeaux; Caroline Robert, Institute Gustave Roussy, Paris; Laurent Mortier, Université de Lille, Institut National de la Santé et de la Recherche Médicale U1189, Centre Hospitalier Universitaire de sa Region Lille, Lille; Thierry Lesimple, Centre Eugène Marquis, Rennes, France; Jacob Schachter, Sheba Medical Center, Tel Hashomer; Tel Aviv University, Tel Aviv, Israel; Kohinoor Dasgupta, Novartis Healthcare, Hyderabad, India; Mark Shilkrut and Eduard Gasal, Novartis Pharmaceuticals Corporation, East Hanover, NJ; and John M. Kirkwood, UPMC Hillman Cancer Center; University of Pittsburgh, Pittsburgh, PA
| | - Dirk Schadendorf
- Axel Hauschild, University Hospital Schleswig-Holstein, Kiel; Dirk Schadendorf, University Hospital Essen, Essen; German Cancer Consortium, Heidelberg, Germany; Reinhard Dummer, University Hospital Zürich Skin Cancer Center, Zürich; Tomas Haas, Novartis AG, Basel, Switzerland; Mario Santinami, Fondazione Istituto Nazionale Tumori, Milan; Mario Mandalà, Papa Giovanni XXIII Cancer Center Hospital, Bergamo; Vanna Chiarion-Sileni, Veneto Institute of Oncology–Istituto di Ricovero e Cura a Carattere Scientifico, Padova, Italy; Victoria Atkinson, Princess Alexandra Hospital; Gallipoli Medical Research Foundation; University of Queensland, Brisbane, Queensland; Andrew Haydon, The Alfred Hospital, Melbourne, Victoria; Richard Kefford, Macquarie University; Westmead Hospital; Richard Kefford and Georgina V. Long, Melanoma Institute Australia; University of Sydney; Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia; James Larkin, Royal Marsden National Health Service Foundation Trust, London; Ruth Plummer, Freeman Hospital and Newcastle University, Newcastle upon Tyne, United Kingdom; Marta Nyakas, Oslo University Hospital, Oslo, Norway; Caroline Dutriaux, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint-André, Bordeaux; Caroline Robert, Institute Gustave Roussy, Paris; Laurent Mortier, Université de Lille, Institut National de la Santé et de la Recherche Médicale U1189, Centre Hospitalier Universitaire de sa Region Lille, Lille; Thierry Lesimple, Centre Eugène Marquis, Rennes, France; Jacob Schachter, Sheba Medical Center, Tel Hashomer; Tel Aviv University, Tel Aviv, Israel; Kohinoor Dasgupta, Novartis Healthcare, Hyderabad, India; Mark Shilkrut and Eduard Gasal, Novartis Pharmaceuticals Corporation, East Hanover, NJ; and John M. Kirkwood, UPMC Hillman Cancer Center; University of Pittsburgh, Pittsburgh, PA
| | - Mario Santinami
- Axel Hauschild, University Hospital Schleswig-Holstein, Kiel; Dirk Schadendorf, University Hospital Essen, Essen; German Cancer Consortium, Heidelberg, Germany; Reinhard Dummer, University Hospital Zürich Skin Cancer Center, Zürich; Tomas Haas, Novartis AG, Basel, Switzerland; Mario Santinami, Fondazione Istituto Nazionale Tumori, Milan; Mario Mandalà, Papa Giovanni XXIII Cancer Center Hospital, Bergamo; Vanna Chiarion-Sileni, Veneto Institute of Oncology–Istituto di Ricovero e Cura a Carattere Scientifico, Padova, Italy; Victoria Atkinson, Princess Alexandra Hospital; Gallipoli Medical Research Foundation; University of Queensland, Brisbane, Queensland; Andrew Haydon, The Alfred Hospital, Melbourne, Victoria; Richard Kefford, Macquarie University; Westmead Hospital; Richard Kefford and Georgina V. Long, Melanoma Institute Australia; University of Sydney; Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia; James Larkin, Royal Marsden National Health Service Foundation Trust, London; Ruth Plummer, Freeman Hospital and Newcastle University, Newcastle upon Tyne, United Kingdom; Marta Nyakas, Oslo University Hospital, Oslo, Norway; Caroline Dutriaux, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint-André, Bordeaux; Caroline Robert, Institute Gustave Roussy, Paris; Laurent Mortier, Université de Lille, Institut National de la Santé et de la Recherche Médicale U1189, Centre Hospitalier Universitaire de sa Region Lille, Lille; Thierry Lesimple, Centre Eugène Marquis, Rennes, France; Jacob Schachter, Sheba Medical Center, Tel Hashomer; Tel Aviv University, Tel Aviv, Israel; Kohinoor Dasgupta, Novartis Healthcare, Hyderabad, India; Mark Shilkrut and Eduard Gasal, Novartis Pharmaceuticals Corporation, East Hanover, NJ; and John M. Kirkwood, UPMC Hillman Cancer Center; University of Pittsburgh, Pittsburgh, PA
| | - Victoria Atkinson
- Axel Hauschild, University Hospital Schleswig-Holstein, Kiel; Dirk Schadendorf, University Hospital Essen, Essen; German Cancer Consortium, Heidelberg, Germany; Reinhard Dummer, University Hospital Zürich Skin Cancer Center, Zürich; Tomas Haas, Novartis AG, Basel, Switzerland; Mario Santinami, Fondazione Istituto Nazionale Tumori, Milan; Mario Mandalà, Papa Giovanni XXIII Cancer Center Hospital, Bergamo; Vanna Chiarion-Sileni, Veneto Institute of Oncology–Istituto di Ricovero e Cura a Carattere Scientifico, Padova, Italy; Victoria Atkinson, Princess Alexandra Hospital; Gallipoli Medical Research Foundation; University of Queensland, Brisbane, Queensland; Andrew Haydon, The Alfred Hospital, Melbourne, Victoria; Richard Kefford, Macquarie University; Westmead Hospital; Richard Kefford and Georgina V. Long, Melanoma Institute Australia; University of Sydney; Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia; James Larkin, Royal Marsden National Health Service Foundation Trust, London; Ruth Plummer, Freeman Hospital and Newcastle University, Newcastle upon Tyne, United Kingdom; Marta Nyakas, Oslo University Hospital, Oslo, Norway; Caroline Dutriaux, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint-André, Bordeaux; Caroline Robert, Institute Gustave Roussy, Paris; Laurent Mortier, Université de Lille, Institut National de la Santé et de la Recherche Médicale U1189, Centre Hospitalier Universitaire de sa Region Lille, Lille; Thierry Lesimple, Centre Eugène Marquis, Rennes, France; Jacob Schachter, Sheba Medical Center, Tel Hashomer; Tel Aviv University, Tel Aviv, Israel; Kohinoor Dasgupta, Novartis Healthcare, Hyderabad, India; Mark Shilkrut and Eduard Gasal, Novartis Pharmaceuticals Corporation, East Hanover, NJ; and John M. Kirkwood, UPMC Hillman Cancer Center; University of Pittsburgh, Pittsburgh, PA
| | - Mario Mandalà
- Axel Hauschild, University Hospital Schleswig-Holstein, Kiel; Dirk Schadendorf, University Hospital Essen, Essen; German Cancer Consortium, Heidelberg, Germany; Reinhard Dummer, University Hospital Zürich Skin Cancer Center, Zürich; Tomas Haas, Novartis AG, Basel, Switzerland; Mario Santinami, Fondazione Istituto Nazionale Tumori, Milan; Mario Mandalà, Papa Giovanni XXIII Cancer Center Hospital, Bergamo; Vanna Chiarion-Sileni, Veneto Institute of Oncology–Istituto di Ricovero e Cura a Carattere Scientifico, Padova, Italy; Victoria Atkinson, Princess Alexandra Hospital; Gallipoli Medical Research Foundation; University of Queensland, Brisbane, Queensland; Andrew Haydon, The Alfred Hospital, Melbourne, Victoria; Richard Kefford, Macquarie University; Westmead Hospital; Richard Kefford and Georgina V. Long, Melanoma Institute Australia; University of Sydney; Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia; James Larkin, Royal Marsden National Health Service Foundation Trust, London; Ruth Plummer, Freeman Hospital and Newcastle University, Newcastle upon Tyne, United Kingdom; Marta Nyakas, Oslo University Hospital, Oslo, Norway; Caroline Dutriaux, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint-André, Bordeaux; Caroline Robert, Institute Gustave Roussy, Paris; Laurent Mortier, Université de Lille, Institut National de la Santé et de la Recherche Médicale U1189, Centre Hospitalier Universitaire de sa Region Lille, Lille; Thierry Lesimple, Centre Eugène Marquis, Rennes, France; Jacob Schachter, Sheba Medical Center, Tel Hashomer; Tel Aviv University, Tel Aviv, Israel; Kohinoor Dasgupta, Novartis Healthcare, Hyderabad, India; Mark Shilkrut and Eduard Gasal, Novartis Pharmaceuticals Corporation, East Hanover, NJ; and John M. Kirkwood, UPMC Hillman Cancer Center; University of Pittsburgh, Pittsburgh, PA
| | - Vanna Chiarion-Sileni
- Axel Hauschild, University Hospital Schleswig-Holstein, Kiel; Dirk Schadendorf, University Hospital Essen, Essen; German Cancer Consortium, Heidelberg, Germany; Reinhard Dummer, University Hospital Zürich Skin Cancer Center, Zürich; Tomas Haas, Novartis AG, Basel, Switzerland; Mario Santinami, Fondazione Istituto Nazionale Tumori, Milan; Mario Mandalà, Papa Giovanni XXIII Cancer Center Hospital, Bergamo; Vanna Chiarion-Sileni, Veneto Institute of Oncology–Istituto di Ricovero e Cura a Carattere Scientifico, Padova, Italy; Victoria Atkinson, Princess Alexandra Hospital; Gallipoli Medical Research Foundation; University of Queensland, Brisbane, Queensland; Andrew Haydon, The Alfred Hospital, Melbourne, Victoria; Richard Kefford, Macquarie University; Westmead Hospital; Richard Kefford and Georgina V. Long, Melanoma Institute Australia; University of Sydney; Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia; James Larkin, Royal Marsden National Health Service Foundation Trust, London; Ruth Plummer, Freeman Hospital and Newcastle University, Newcastle upon Tyne, United Kingdom; Marta Nyakas, Oslo University Hospital, Oslo, Norway; Caroline Dutriaux, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint-André, Bordeaux; Caroline Robert, Institute Gustave Roussy, Paris; Laurent Mortier, Université de Lille, Institut National de la Santé et de la Recherche Médicale U1189, Centre Hospitalier Universitaire de sa Region Lille, Lille; Thierry Lesimple, Centre Eugène Marquis, Rennes, France; Jacob Schachter, Sheba Medical Center, Tel Hashomer; Tel Aviv University, Tel Aviv, Israel; Kohinoor Dasgupta, Novartis Healthcare, Hyderabad, India; Mark Shilkrut and Eduard Gasal, Novartis Pharmaceuticals Corporation, East Hanover, NJ; and John M. Kirkwood, UPMC Hillman Cancer Center; University of Pittsburgh, Pittsburgh, PA
| | - James Larkin
- Axel Hauschild, University Hospital Schleswig-Holstein, Kiel; Dirk Schadendorf, University Hospital Essen, Essen; German Cancer Consortium, Heidelberg, Germany; Reinhard Dummer, University Hospital Zürich Skin Cancer Center, Zürich; Tomas Haas, Novartis AG, Basel, Switzerland; Mario Santinami, Fondazione Istituto Nazionale Tumori, Milan; Mario Mandalà, Papa Giovanni XXIII Cancer Center Hospital, Bergamo; Vanna Chiarion-Sileni, Veneto Institute of Oncology–Istituto di Ricovero e Cura a Carattere Scientifico, Padova, Italy; Victoria Atkinson, Princess Alexandra Hospital; Gallipoli Medical Research Foundation; University of Queensland, Brisbane, Queensland; Andrew Haydon, The Alfred Hospital, Melbourne, Victoria; Richard Kefford, Macquarie University; Westmead Hospital; Richard Kefford and Georgina V. Long, Melanoma Institute Australia; University of Sydney; Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia; James Larkin, Royal Marsden National Health Service Foundation Trust, London; Ruth Plummer, Freeman Hospital and Newcastle University, Newcastle upon Tyne, United Kingdom; Marta Nyakas, Oslo University Hospital, Oslo, Norway; Caroline Dutriaux, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint-André, Bordeaux; Caroline Robert, Institute Gustave Roussy, Paris; Laurent Mortier, Université de Lille, Institut National de la Santé et de la Recherche Médicale U1189, Centre Hospitalier Universitaire de sa Region Lille, Lille; Thierry Lesimple, Centre Eugène Marquis, Rennes, France; Jacob Schachter, Sheba Medical Center, Tel Hashomer; Tel Aviv University, Tel Aviv, Israel; Kohinoor Dasgupta, Novartis Healthcare, Hyderabad, India; Mark Shilkrut and Eduard Gasal, Novartis Pharmaceuticals Corporation, East Hanover, NJ; and John M. Kirkwood, UPMC Hillman Cancer Center; University of Pittsburgh, Pittsburgh, PA
| | - Marta Nyakas
- Axel Hauschild, University Hospital Schleswig-Holstein, Kiel; Dirk Schadendorf, University Hospital Essen, Essen; German Cancer Consortium, Heidelberg, Germany; Reinhard Dummer, University Hospital Zürich Skin Cancer Center, Zürich; Tomas Haas, Novartis AG, Basel, Switzerland; Mario Santinami, Fondazione Istituto Nazionale Tumori, Milan; Mario Mandalà, Papa Giovanni XXIII Cancer Center Hospital, Bergamo; Vanna Chiarion-Sileni, Veneto Institute of Oncology–Istituto di Ricovero e Cura a Carattere Scientifico, Padova, Italy; Victoria Atkinson, Princess Alexandra Hospital; Gallipoli Medical Research Foundation; University of Queensland, Brisbane, Queensland; Andrew Haydon, The Alfred Hospital, Melbourne, Victoria; Richard Kefford, Macquarie University; Westmead Hospital; Richard Kefford and Georgina V. Long, Melanoma Institute Australia; University of Sydney; Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia; James Larkin, Royal Marsden National Health Service Foundation Trust, London; Ruth Plummer, Freeman Hospital and Newcastle University, Newcastle upon Tyne, United Kingdom; Marta Nyakas, Oslo University Hospital, Oslo, Norway; Caroline Dutriaux, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint-André, Bordeaux; Caroline Robert, Institute Gustave Roussy, Paris; Laurent Mortier, Université de Lille, Institut National de la Santé et de la Recherche Médicale U1189, Centre Hospitalier Universitaire de sa Region Lille, Lille; Thierry Lesimple, Centre Eugène Marquis, Rennes, France; Jacob Schachter, Sheba Medical Center, Tel Hashomer; Tel Aviv University, Tel Aviv, Israel; Kohinoor Dasgupta, Novartis Healthcare, Hyderabad, India; Mark Shilkrut and Eduard Gasal, Novartis Pharmaceuticals Corporation, East Hanover, NJ; and John M. Kirkwood, UPMC Hillman Cancer Center; University of Pittsburgh, Pittsburgh, PA
| | - Caroline Dutriaux
- Axel Hauschild, University Hospital Schleswig-Holstein, Kiel; Dirk Schadendorf, University Hospital Essen, Essen; German Cancer Consortium, Heidelberg, Germany; Reinhard Dummer, University Hospital Zürich Skin Cancer Center, Zürich; Tomas Haas, Novartis AG, Basel, Switzerland; Mario Santinami, Fondazione Istituto Nazionale Tumori, Milan; Mario Mandalà, Papa Giovanni XXIII Cancer Center Hospital, Bergamo; Vanna Chiarion-Sileni, Veneto Institute of Oncology–Istituto di Ricovero e Cura a Carattere Scientifico, Padova, Italy; Victoria Atkinson, Princess Alexandra Hospital; Gallipoli Medical Research Foundation; University of Queensland, Brisbane, Queensland; Andrew Haydon, The Alfred Hospital, Melbourne, Victoria; Richard Kefford, Macquarie University; Westmead Hospital; Richard Kefford and Georgina V. Long, Melanoma Institute Australia; University of Sydney; Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia; James Larkin, Royal Marsden National Health Service Foundation Trust, London; Ruth Plummer, Freeman Hospital and Newcastle University, Newcastle upon Tyne, United Kingdom; Marta Nyakas, Oslo University Hospital, Oslo, Norway; Caroline Dutriaux, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint-André, Bordeaux; Caroline Robert, Institute Gustave Roussy, Paris; Laurent Mortier, Université de Lille, Institut National de la Santé et de la Recherche Médicale U1189, Centre Hospitalier Universitaire de sa Region Lille, Lille; Thierry Lesimple, Centre Eugène Marquis, Rennes, France; Jacob Schachter, Sheba Medical Center, Tel Hashomer; Tel Aviv University, Tel Aviv, Israel; Kohinoor Dasgupta, Novartis Healthcare, Hyderabad, India; Mark Shilkrut and Eduard Gasal, Novartis Pharmaceuticals Corporation, East Hanover, NJ; and John M. Kirkwood, UPMC Hillman Cancer Center; University of Pittsburgh, Pittsburgh, PA
| | - Andrew Haydon
- Axel Hauschild, University Hospital Schleswig-Holstein, Kiel; Dirk Schadendorf, University Hospital Essen, Essen; German Cancer Consortium, Heidelberg, Germany; Reinhard Dummer, University Hospital Zürich Skin Cancer Center, Zürich; Tomas Haas, Novartis AG, Basel, Switzerland; Mario Santinami, Fondazione Istituto Nazionale Tumori, Milan; Mario Mandalà, Papa Giovanni XXIII Cancer Center Hospital, Bergamo; Vanna Chiarion-Sileni, Veneto Institute of Oncology–Istituto di Ricovero e Cura a Carattere Scientifico, Padova, Italy; Victoria Atkinson, Princess Alexandra Hospital; Gallipoli Medical Research Foundation; University of Queensland, Brisbane, Queensland; Andrew Haydon, The Alfred Hospital, Melbourne, Victoria; Richard Kefford, Macquarie University; Westmead Hospital; Richard Kefford and Georgina V. Long, Melanoma Institute Australia; University of Sydney; Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia; James Larkin, Royal Marsden National Health Service Foundation Trust, London; Ruth Plummer, Freeman Hospital and Newcastle University, Newcastle upon Tyne, United Kingdom; Marta Nyakas, Oslo University Hospital, Oslo, Norway; Caroline Dutriaux, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint-André, Bordeaux; Caroline Robert, Institute Gustave Roussy, Paris; Laurent Mortier, Université de Lille, Institut National de la Santé et de la Recherche Médicale U1189, Centre Hospitalier Universitaire de sa Region Lille, Lille; Thierry Lesimple, Centre Eugène Marquis, Rennes, France; Jacob Schachter, Sheba Medical Center, Tel Hashomer; Tel Aviv University, Tel Aviv, Israel; Kohinoor Dasgupta, Novartis Healthcare, Hyderabad, India; Mark Shilkrut and Eduard Gasal, Novartis Pharmaceuticals Corporation, East Hanover, NJ; and John M. Kirkwood, UPMC Hillman Cancer Center; University of Pittsburgh, Pittsburgh, PA
| | - Caroline Robert
- Axel Hauschild, University Hospital Schleswig-Holstein, Kiel; Dirk Schadendorf, University Hospital Essen, Essen; German Cancer Consortium, Heidelberg, Germany; Reinhard Dummer, University Hospital Zürich Skin Cancer Center, Zürich; Tomas Haas, Novartis AG, Basel, Switzerland; Mario Santinami, Fondazione Istituto Nazionale Tumori, Milan; Mario Mandalà, Papa Giovanni XXIII Cancer Center Hospital, Bergamo; Vanna Chiarion-Sileni, Veneto Institute of Oncology–Istituto di Ricovero e Cura a Carattere Scientifico, Padova, Italy; Victoria Atkinson, Princess Alexandra Hospital; Gallipoli Medical Research Foundation; University of Queensland, Brisbane, Queensland; Andrew Haydon, The Alfred Hospital, Melbourne, Victoria; Richard Kefford, Macquarie University; Westmead Hospital; Richard Kefford and Georgina V. Long, Melanoma Institute Australia; University of Sydney; Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia; James Larkin, Royal Marsden National Health Service Foundation Trust, London; Ruth Plummer, Freeman Hospital and Newcastle University, Newcastle upon Tyne, United Kingdom; Marta Nyakas, Oslo University Hospital, Oslo, Norway; Caroline Dutriaux, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint-André, Bordeaux; Caroline Robert, Institute Gustave Roussy, Paris; Laurent Mortier, Université de Lille, Institut National de la Santé et de la Recherche Médicale U1189, Centre Hospitalier Universitaire de sa Region Lille, Lille; Thierry Lesimple, Centre Eugène Marquis, Rennes, France; Jacob Schachter, Sheba Medical Center, Tel Hashomer; Tel Aviv University, Tel Aviv, Israel; Kohinoor Dasgupta, Novartis Healthcare, Hyderabad, India; Mark Shilkrut and Eduard Gasal, Novartis Pharmaceuticals Corporation, East Hanover, NJ; and John M. Kirkwood, UPMC Hillman Cancer Center; University of Pittsburgh, Pittsburgh, PA
| | - Laurent Mortier
- Axel Hauschild, University Hospital Schleswig-Holstein, Kiel; Dirk Schadendorf, University Hospital Essen, Essen; German Cancer Consortium, Heidelberg, Germany; Reinhard Dummer, University Hospital Zürich Skin Cancer Center, Zürich; Tomas Haas, Novartis AG, Basel, Switzerland; Mario Santinami, Fondazione Istituto Nazionale Tumori, Milan; Mario Mandalà, Papa Giovanni XXIII Cancer Center Hospital, Bergamo; Vanna Chiarion-Sileni, Veneto Institute of Oncology–Istituto di Ricovero e Cura a Carattere Scientifico, Padova, Italy; Victoria Atkinson, Princess Alexandra Hospital; Gallipoli Medical Research Foundation; University of Queensland, Brisbane, Queensland; Andrew Haydon, The Alfred Hospital, Melbourne, Victoria; Richard Kefford, Macquarie University; Westmead Hospital; Richard Kefford and Georgina V. Long, Melanoma Institute Australia; University of Sydney; Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia; James Larkin, Royal Marsden National Health Service Foundation Trust, London; Ruth Plummer, Freeman Hospital and Newcastle University, Newcastle upon Tyne, United Kingdom; Marta Nyakas, Oslo University Hospital, Oslo, Norway; Caroline Dutriaux, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint-André, Bordeaux; Caroline Robert, Institute Gustave Roussy, Paris; Laurent Mortier, Université de Lille, Institut National de la Santé et de la Recherche Médicale U1189, Centre Hospitalier Universitaire de sa Region Lille, Lille; Thierry Lesimple, Centre Eugène Marquis, Rennes, France; Jacob Schachter, Sheba Medical Center, Tel Hashomer; Tel Aviv University, Tel Aviv, Israel; Kohinoor Dasgupta, Novartis Healthcare, Hyderabad, India; Mark Shilkrut and Eduard Gasal, Novartis Pharmaceuticals Corporation, East Hanover, NJ; and John M. Kirkwood, UPMC Hillman Cancer Center; University of Pittsburgh, Pittsburgh, PA
| | - Jacob Schachter
- Axel Hauschild, University Hospital Schleswig-Holstein, Kiel; Dirk Schadendorf, University Hospital Essen, Essen; German Cancer Consortium, Heidelberg, Germany; Reinhard Dummer, University Hospital Zürich Skin Cancer Center, Zürich; Tomas Haas, Novartis AG, Basel, Switzerland; Mario Santinami, Fondazione Istituto Nazionale Tumori, Milan; Mario Mandalà, Papa Giovanni XXIII Cancer Center Hospital, Bergamo; Vanna Chiarion-Sileni, Veneto Institute of Oncology–Istituto di Ricovero e Cura a Carattere Scientifico, Padova, Italy; Victoria Atkinson, Princess Alexandra Hospital; Gallipoli Medical Research Foundation; University of Queensland, Brisbane, Queensland; Andrew Haydon, The Alfred Hospital, Melbourne, Victoria; Richard Kefford, Macquarie University; Westmead Hospital; Richard Kefford and Georgina V. Long, Melanoma Institute Australia; University of Sydney; Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia; James Larkin, Royal Marsden National Health Service Foundation Trust, London; Ruth Plummer, Freeman Hospital and Newcastle University, Newcastle upon Tyne, United Kingdom; Marta Nyakas, Oslo University Hospital, Oslo, Norway; Caroline Dutriaux, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint-André, Bordeaux; Caroline Robert, Institute Gustave Roussy, Paris; Laurent Mortier, Université de Lille, Institut National de la Santé et de la Recherche Médicale U1189, Centre Hospitalier Universitaire de sa Region Lille, Lille; Thierry Lesimple, Centre Eugène Marquis, Rennes, France; Jacob Schachter, Sheba Medical Center, Tel Hashomer; Tel Aviv University, Tel Aviv, Israel; Kohinoor Dasgupta, Novartis Healthcare, Hyderabad, India; Mark Shilkrut and Eduard Gasal, Novartis Pharmaceuticals Corporation, East Hanover, NJ; and John M. Kirkwood, UPMC Hillman Cancer Center; University of Pittsburgh, Pittsburgh, PA
| | - Thierry Lesimple
- Axel Hauschild, University Hospital Schleswig-Holstein, Kiel; Dirk Schadendorf, University Hospital Essen, Essen; German Cancer Consortium, Heidelberg, Germany; Reinhard Dummer, University Hospital Zürich Skin Cancer Center, Zürich; Tomas Haas, Novartis AG, Basel, Switzerland; Mario Santinami, Fondazione Istituto Nazionale Tumori, Milan; Mario Mandalà, Papa Giovanni XXIII Cancer Center Hospital, Bergamo; Vanna Chiarion-Sileni, Veneto Institute of Oncology–Istituto di Ricovero e Cura a Carattere Scientifico, Padova, Italy; Victoria Atkinson, Princess Alexandra Hospital; Gallipoli Medical Research Foundation; University of Queensland, Brisbane, Queensland; Andrew Haydon, The Alfred Hospital, Melbourne, Victoria; Richard Kefford, Macquarie University; Westmead Hospital; Richard Kefford and Georgina V. Long, Melanoma Institute Australia; University of Sydney; Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia; James Larkin, Royal Marsden National Health Service Foundation Trust, London; Ruth Plummer, Freeman Hospital and Newcastle University, Newcastle upon Tyne, United Kingdom; Marta Nyakas, Oslo University Hospital, Oslo, Norway; Caroline Dutriaux, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint-André, Bordeaux; Caroline Robert, Institute Gustave Roussy, Paris; Laurent Mortier, Université de Lille, Institut National de la Santé et de la Recherche Médicale U1189, Centre Hospitalier Universitaire de sa Region Lille, Lille; Thierry Lesimple, Centre Eugène Marquis, Rennes, France; Jacob Schachter, Sheba Medical Center, Tel Hashomer; Tel Aviv University, Tel Aviv, Israel; Kohinoor Dasgupta, Novartis Healthcare, Hyderabad, India; Mark Shilkrut and Eduard Gasal, Novartis Pharmaceuticals Corporation, East Hanover, NJ; and John M. Kirkwood, UPMC Hillman Cancer Center; University of Pittsburgh, Pittsburgh, PA
| | - Ruth Plummer
- Axel Hauschild, University Hospital Schleswig-Holstein, Kiel; Dirk Schadendorf, University Hospital Essen, Essen; German Cancer Consortium, Heidelberg, Germany; Reinhard Dummer, University Hospital Zürich Skin Cancer Center, Zürich; Tomas Haas, Novartis AG, Basel, Switzerland; Mario Santinami, Fondazione Istituto Nazionale Tumori, Milan; Mario Mandalà, Papa Giovanni XXIII Cancer Center Hospital, Bergamo; Vanna Chiarion-Sileni, Veneto Institute of Oncology–Istituto di Ricovero e Cura a Carattere Scientifico, Padova, Italy; Victoria Atkinson, Princess Alexandra Hospital; Gallipoli Medical Research Foundation; University of Queensland, Brisbane, Queensland; Andrew Haydon, The Alfred Hospital, Melbourne, Victoria; Richard Kefford, Macquarie University; Westmead Hospital; Richard Kefford and Georgina V. Long, Melanoma Institute Australia; University of Sydney; Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia; James Larkin, Royal Marsden National Health Service Foundation Trust, London; Ruth Plummer, Freeman Hospital and Newcastle University, Newcastle upon Tyne, United Kingdom; Marta Nyakas, Oslo University Hospital, Oslo, Norway; Caroline Dutriaux, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint-André, Bordeaux; Caroline Robert, Institute Gustave Roussy, Paris; Laurent Mortier, Université de Lille, Institut National de la Santé et de la Recherche Médicale U1189, Centre Hospitalier Universitaire de sa Region Lille, Lille; Thierry Lesimple, Centre Eugène Marquis, Rennes, France; Jacob Schachter, Sheba Medical Center, Tel Hashomer; Tel Aviv University, Tel Aviv, Israel; Kohinoor Dasgupta, Novartis Healthcare, Hyderabad, India; Mark Shilkrut and Eduard Gasal, Novartis Pharmaceuticals Corporation, East Hanover, NJ; and John M. Kirkwood, UPMC Hillman Cancer Center; University of Pittsburgh, Pittsburgh, PA
| | - Kohinoor Dasgupta
- Axel Hauschild, University Hospital Schleswig-Holstein, Kiel; Dirk Schadendorf, University Hospital Essen, Essen; German Cancer Consortium, Heidelberg, Germany; Reinhard Dummer, University Hospital Zürich Skin Cancer Center, Zürich; Tomas Haas, Novartis AG, Basel, Switzerland; Mario Santinami, Fondazione Istituto Nazionale Tumori, Milan; Mario Mandalà, Papa Giovanni XXIII Cancer Center Hospital, Bergamo; Vanna Chiarion-Sileni, Veneto Institute of Oncology–Istituto di Ricovero e Cura a Carattere Scientifico, Padova, Italy; Victoria Atkinson, Princess Alexandra Hospital; Gallipoli Medical Research Foundation; University of Queensland, Brisbane, Queensland; Andrew Haydon, The Alfred Hospital, Melbourne, Victoria; Richard Kefford, Macquarie University; Westmead Hospital; Richard Kefford and Georgina V. Long, Melanoma Institute Australia; University of Sydney; Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia; James Larkin, Royal Marsden National Health Service Foundation Trust, London; Ruth Plummer, Freeman Hospital and Newcastle University, Newcastle upon Tyne, United Kingdom; Marta Nyakas, Oslo University Hospital, Oslo, Norway; Caroline Dutriaux, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint-André, Bordeaux; Caroline Robert, Institute Gustave Roussy, Paris; Laurent Mortier, Université de Lille, Institut National de la Santé et de la Recherche Médicale U1189, Centre Hospitalier Universitaire de sa Region Lille, Lille; Thierry Lesimple, Centre Eugène Marquis, Rennes, France; Jacob Schachter, Sheba Medical Center, Tel Hashomer; Tel Aviv University, Tel Aviv, Israel; Kohinoor Dasgupta, Novartis Healthcare, Hyderabad, India; Mark Shilkrut and Eduard Gasal, Novartis Pharmaceuticals Corporation, East Hanover, NJ; and John M. Kirkwood, UPMC Hillman Cancer Center; University of Pittsburgh, Pittsburgh, PA
| | - Tomas Haas
- Axel Hauschild, University Hospital Schleswig-Holstein, Kiel; Dirk Schadendorf, University Hospital Essen, Essen; German Cancer Consortium, Heidelberg, Germany; Reinhard Dummer, University Hospital Zürich Skin Cancer Center, Zürich; Tomas Haas, Novartis AG, Basel, Switzerland; Mario Santinami, Fondazione Istituto Nazionale Tumori, Milan; Mario Mandalà, Papa Giovanni XXIII Cancer Center Hospital, Bergamo; Vanna Chiarion-Sileni, Veneto Institute of Oncology–Istituto di Ricovero e Cura a Carattere Scientifico, Padova, Italy; Victoria Atkinson, Princess Alexandra Hospital; Gallipoli Medical Research Foundation; University of Queensland, Brisbane, Queensland; Andrew Haydon, The Alfred Hospital, Melbourne, Victoria; Richard Kefford, Macquarie University; Westmead Hospital; Richard Kefford and Georgina V. Long, Melanoma Institute Australia; University of Sydney; Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia; James Larkin, Royal Marsden National Health Service Foundation Trust, London; Ruth Plummer, Freeman Hospital and Newcastle University, Newcastle upon Tyne, United Kingdom; Marta Nyakas, Oslo University Hospital, Oslo, Norway; Caroline Dutriaux, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint-André, Bordeaux; Caroline Robert, Institute Gustave Roussy, Paris; Laurent Mortier, Université de Lille, Institut National de la Santé et de la Recherche Médicale U1189, Centre Hospitalier Universitaire de sa Region Lille, Lille; Thierry Lesimple, Centre Eugène Marquis, Rennes, France; Jacob Schachter, Sheba Medical Center, Tel Hashomer; Tel Aviv University, Tel Aviv, Israel; Kohinoor Dasgupta, Novartis Healthcare, Hyderabad, India; Mark Shilkrut and Eduard Gasal, Novartis Pharmaceuticals Corporation, East Hanover, NJ; and John M. Kirkwood, UPMC Hillman Cancer Center; University of Pittsburgh, Pittsburgh, PA
| | - Mark Shilkrut
- Axel Hauschild, University Hospital Schleswig-Holstein, Kiel; Dirk Schadendorf, University Hospital Essen, Essen; German Cancer Consortium, Heidelberg, Germany; Reinhard Dummer, University Hospital Zürich Skin Cancer Center, Zürich; Tomas Haas, Novartis AG, Basel, Switzerland; Mario Santinami, Fondazione Istituto Nazionale Tumori, Milan; Mario Mandalà, Papa Giovanni XXIII Cancer Center Hospital, Bergamo; Vanna Chiarion-Sileni, Veneto Institute of Oncology–Istituto di Ricovero e Cura a Carattere Scientifico, Padova, Italy; Victoria Atkinson, Princess Alexandra Hospital; Gallipoli Medical Research Foundation; University of Queensland, Brisbane, Queensland; Andrew Haydon, The Alfred Hospital, Melbourne, Victoria; Richard Kefford, Macquarie University; Westmead Hospital; Richard Kefford and Georgina V. Long, Melanoma Institute Australia; University of Sydney; Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia; James Larkin, Royal Marsden National Health Service Foundation Trust, London; Ruth Plummer, Freeman Hospital and Newcastle University, Newcastle upon Tyne, United Kingdom; Marta Nyakas, Oslo University Hospital, Oslo, Norway; Caroline Dutriaux, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint-André, Bordeaux; Caroline Robert, Institute Gustave Roussy, Paris; Laurent Mortier, Université de Lille, Institut National de la Santé et de la Recherche Médicale U1189, Centre Hospitalier Universitaire de sa Region Lille, Lille; Thierry Lesimple, Centre Eugène Marquis, Rennes, France; Jacob Schachter, Sheba Medical Center, Tel Hashomer; Tel Aviv University, Tel Aviv, Israel; Kohinoor Dasgupta, Novartis Healthcare, Hyderabad, India; Mark Shilkrut and Eduard Gasal, Novartis Pharmaceuticals Corporation, East Hanover, NJ; and John M. Kirkwood, UPMC Hillman Cancer Center; University of Pittsburgh, Pittsburgh, PA
| | - Eduard Gasal
- Axel Hauschild, University Hospital Schleswig-Holstein, Kiel; Dirk Schadendorf, University Hospital Essen, Essen; German Cancer Consortium, Heidelberg, Germany; Reinhard Dummer, University Hospital Zürich Skin Cancer Center, Zürich; Tomas Haas, Novartis AG, Basel, Switzerland; Mario Santinami, Fondazione Istituto Nazionale Tumori, Milan; Mario Mandalà, Papa Giovanni XXIII Cancer Center Hospital, Bergamo; Vanna Chiarion-Sileni, Veneto Institute of Oncology–Istituto di Ricovero e Cura a Carattere Scientifico, Padova, Italy; Victoria Atkinson, Princess Alexandra Hospital; Gallipoli Medical Research Foundation; University of Queensland, Brisbane, Queensland; Andrew Haydon, The Alfred Hospital, Melbourne, Victoria; Richard Kefford, Macquarie University; Westmead Hospital; Richard Kefford and Georgina V. Long, Melanoma Institute Australia; University of Sydney; Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia; James Larkin, Royal Marsden National Health Service Foundation Trust, London; Ruth Plummer, Freeman Hospital and Newcastle University, Newcastle upon Tyne, United Kingdom; Marta Nyakas, Oslo University Hospital, Oslo, Norway; Caroline Dutriaux, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint-André, Bordeaux; Caroline Robert, Institute Gustave Roussy, Paris; Laurent Mortier, Université de Lille, Institut National de la Santé et de la Recherche Médicale U1189, Centre Hospitalier Universitaire de sa Region Lille, Lille; Thierry Lesimple, Centre Eugène Marquis, Rennes, France; Jacob Schachter, Sheba Medical Center, Tel Hashomer; Tel Aviv University, Tel Aviv, Israel; Kohinoor Dasgupta, Novartis Healthcare, Hyderabad, India; Mark Shilkrut and Eduard Gasal, Novartis Pharmaceuticals Corporation, East Hanover, NJ; and John M. Kirkwood, UPMC Hillman Cancer Center; University of Pittsburgh, Pittsburgh, PA
| | - Richard Kefford
- Axel Hauschild, University Hospital Schleswig-Holstein, Kiel; Dirk Schadendorf, University Hospital Essen, Essen; German Cancer Consortium, Heidelberg, Germany; Reinhard Dummer, University Hospital Zürich Skin Cancer Center, Zürich; Tomas Haas, Novartis AG, Basel, Switzerland; Mario Santinami, Fondazione Istituto Nazionale Tumori, Milan; Mario Mandalà, Papa Giovanni XXIII Cancer Center Hospital, Bergamo; Vanna Chiarion-Sileni, Veneto Institute of Oncology–Istituto di Ricovero e Cura a Carattere Scientifico, Padova, Italy; Victoria Atkinson, Princess Alexandra Hospital; Gallipoli Medical Research Foundation; University of Queensland, Brisbane, Queensland; Andrew Haydon, The Alfred Hospital, Melbourne, Victoria; Richard Kefford, Macquarie University; Westmead Hospital; Richard Kefford and Georgina V. Long, Melanoma Institute Australia; University of Sydney; Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia; James Larkin, Royal Marsden National Health Service Foundation Trust, London; Ruth Plummer, Freeman Hospital and Newcastle University, Newcastle upon Tyne, United Kingdom; Marta Nyakas, Oslo University Hospital, Oslo, Norway; Caroline Dutriaux, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint-André, Bordeaux; Caroline Robert, Institute Gustave Roussy, Paris; Laurent Mortier, Université de Lille, Institut National de la Santé et de la Recherche Médicale U1189, Centre Hospitalier Universitaire de sa Region Lille, Lille; Thierry Lesimple, Centre Eugène Marquis, Rennes, France; Jacob Schachter, Sheba Medical Center, Tel Hashomer; Tel Aviv University, Tel Aviv, Israel; Kohinoor Dasgupta, Novartis Healthcare, Hyderabad, India; Mark Shilkrut and Eduard Gasal, Novartis Pharmaceuticals Corporation, East Hanover, NJ; and John M. Kirkwood, UPMC Hillman Cancer Center; University of Pittsburgh, Pittsburgh, PA
| | - John M. Kirkwood
- Axel Hauschild, University Hospital Schleswig-Holstein, Kiel; Dirk Schadendorf, University Hospital Essen, Essen; German Cancer Consortium, Heidelberg, Germany; Reinhard Dummer, University Hospital Zürich Skin Cancer Center, Zürich; Tomas Haas, Novartis AG, Basel, Switzerland; Mario Santinami, Fondazione Istituto Nazionale Tumori, Milan; Mario Mandalà, Papa Giovanni XXIII Cancer Center Hospital, Bergamo; Vanna Chiarion-Sileni, Veneto Institute of Oncology–Istituto di Ricovero e Cura a Carattere Scientifico, Padova, Italy; Victoria Atkinson, Princess Alexandra Hospital; Gallipoli Medical Research Foundation; University of Queensland, Brisbane, Queensland; Andrew Haydon, The Alfred Hospital, Melbourne, Victoria; Richard Kefford, Macquarie University; Westmead Hospital; Richard Kefford and Georgina V. Long, Melanoma Institute Australia; University of Sydney; Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia; James Larkin, Royal Marsden National Health Service Foundation Trust, London; Ruth Plummer, Freeman Hospital and Newcastle University, Newcastle upon Tyne, United Kingdom; Marta Nyakas, Oslo University Hospital, Oslo, Norway; Caroline Dutriaux, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint-André, Bordeaux; Caroline Robert, Institute Gustave Roussy, Paris; Laurent Mortier, Université de Lille, Institut National de la Santé et de la Recherche Médicale U1189, Centre Hospitalier Universitaire de sa Region Lille, Lille; Thierry Lesimple, Centre Eugène Marquis, Rennes, France; Jacob Schachter, Sheba Medical Center, Tel Hashomer; Tel Aviv University, Tel Aviv, Israel; Kohinoor Dasgupta, Novartis Healthcare, Hyderabad, India; Mark Shilkrut and Eduard Gasal, Novartis Pharmaceuticals Corporation, East Hanover, NJ; and John M. Kirkwood, UPMC Hillman Cancer Center; University of Pittsburgh, Pittsburgh, PA
| | - Georgina V. Long
- Axel Hauschild, University Hospital Schleswig-Holstein, Kiel; Dirk Schadendorf, University Hospital Essen, Essen; German Cancer Consortium, Heidelberg, Germany; Reinhard Dummer, University Hospital Zürich Skin Cancer Center, Zürich; Tomas Haas, Novartis AG, Basel, Switzerland; Mario Santinami, Fondazione Istituto Nazionale Tumori, Milan; Mario Mandalà, Papa Giovanni XXIII Cancer Center Hospital, Bergamo; Vanna Chiarion-Sileni, Veneto Institute of Oncology–Istituto di Ricovero e Cura a Carattere Scientifico, Padova, Italy; Victoria Atkinson, Princess Alexandra Hospital; Gallipoli Medical Research Foundation; University of Queensland, Brisbane, Queensland; Andrew Haydon, The Alfred Hospital, Melbourne, Victoria; Richard Kefford, Macquarie University; Westmead Hospital; Richard Kefford and Georgina V. Long, Melanoma Institute Australia; University of Sydney; Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia; James Larkin, Royal Marsden National Health Service Foundation Trust, London; Ruth Plummer, Freeman Hospital and Newcastle University, Newcastle upon Tyne, United Kingdom; Marta Nyakas, Oslo University Hospital, Oslo, Norway; Caroline Dutriaux, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint-André, Bordeaux; Caroline Robert, Institute Gustave Roussy, Paris; Laurent Mortier, Université de Lille, Institut National de la Santé et de la Recherche Médicale U1189, Centre Hospitalier Universitaire de sa Region Lille, Lille; Thierry Lesimple, Centre Eugène Marquis, Rennes, France; Jacob Schachter, Sheba Medical Center, Tel Hashomer; Tel Aviv University, Tel Aviv, Israel; Kohinoor Dasgupta, Novartis Healthcare, Hyderabad, India; Mark Shilkrut and Eduard Gasal, Novartis Pharmaceuticals Corporation, East Hanover, NJ; and John M. Kirkwood, UPMC Hillman Cancer Center; University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
754
|
Sahni S, Valecha G, Sahni A. Role of Anti-PD-1 Antibodies in Advanced Melanoma: The Era of Immunotherapy. Cureus 2018; 10:e3700. [PMID: 30788189 PMCID: PMC6372252 DOI: 10.7759/cureus.3700] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 11/14/2018] [Indexed: 01/23/2023] Open
Abstract
Advanced melanoma is an aggressive skin cancer characterized by poor survival rates and response to cytotoxic chemotherapy. Immune checkpoint inhibitors are novel agents capable of utilizing one's own immune system to bring about the tumor destruction. Nivolumab and pembrolizumab are fully humanized anti-PD-1 monoclonal antibodies that have shown significant anti-tumor activity in a variety of cancers including melanoma and have significantly improved the survival outcomes in patients with advanced melanoma. In this updated review article, we will discuss the outcomes of various clinical trials evaluating the efficacy and safety of these agents. We will also briefly discuss their mechanism of action and adverse effects.
Collapse
Affiliation(s)
- Sakshi Sahni
- Internal Medicine, University of Illinois at Chicago College of Medicine, Chicago, USA
| | - Gautam Valecha
- Hematology-Oncology, Staten Island University Hospital, Staten Island, USA
| | - Ankit Sahni
- Miscellaneous, Ross University School of Medicine, Knoxville, USA
| |
Collapse
|
755
|
Gajiwala S, Torgeson A, Garrido-Laguna I, Kinsey C, Lloyd S. Combination immunotherapy and radiation therapy strategies for pancreatic cancer-targeting multiple steps in the cancer immunity cycle. J Gastrointest Oncol 2018; 9:1014-1026. [PMID: 30603120 PMCID: PMC6286952 DOI: 10.21037/jgo.2018.05.16] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 05/16/2018] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a deadly disease, with its mortality rate approaching its incidence rate every year. Accordingly, much interest has been generated in harnessing the immune system in order to improve survival outcomes for these patients. Pancreatic cancer is not thought to be as immunogenic as other cancers that have seen promising results with immune checkpoint inhibitors alone, therefore likely several targets within the cancer-immunity cycle will need to be employed for successful treatment. We sought to investigate both the current state of the field in immunotherapy in PDAC with a special emphasis on combined approaches with radiation therapy (RT). We also summarized ongoing clinical trials that are examining the use of radiotherapy with other immune-stimulating agents in the treatment of PDAC. A PubMed and clinicaltrials.gov search was conducted using the following search terms, either alone or in combination: "pancreatic cancer", "immunotherapy", and "abscopal effect". Open clinical trials were reviewed and included if they involved both RT and other immune-stimulating agents. Pancreatic cancers tend to reside within immune-suppressive tumor microenvironments (TME), express PD-L1, and secrete several immuno-suppressive agents, such as TGF-B, IL-10, indoleamine 2,3-dioxygenase, galectin-1. Whole-cell vaccine therapies, peptide and protein vaccines, dendritic cell vaccines, and vaccines with micro-organisms have been investigated by themselves with promising results. Open clinical trials are currently investigating the use of these vaccines, which increase antigen presentation, with treatments that stimulate release of tumor antigens including RT. There are currently at least 21 open clinical trials investigating the combination of RT with other immune-stimulating agents. The combination of RT and immunotherapy may be a promising avenue for PDAC treatment and deserves further research.
Collapse
Affiliation(s)
- Snehal Gajiwala
- University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Anna Torgeson
- Department of Radiation Oncology, Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Ignacio Garrido-Laguna
- Department of Internal Medicine, Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Conan Kinsey
- Department of Internal Medicine, Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Shane Lloyd
- Department of Radiation Oncology, Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| |
Collapse
|
756
|
Lyth J. Conditional recurrence-free survival in patients with primary stage I-II cutaneous malignant melanoma - a population-based study. Melanoma Res 2018; 28:637-640. [PMID: 29994850 DOI: 10.1097/cmr.0000000000000479] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Conditional survival in patients with localized primary cutaneous malignant melanoma (CMM) is well described. However, conditional recurrence-free survival (RFS) has not been investigated before. The aim of this study was to determine conditional RFS and test for time dependency in prognostic factors in patients with localized stage I-II CMM. This study included 1437 CMM patients registered in one region of Sweden during 1999-2012 followed up through 31 December 2012. To identify first recurrence of CMM disease, data from a care data warehouse, the pathology and radiology department registries were used. Patients were also followed through a Census Register and the National Cause of Death Register. The time-dependent risk of recurrence was analysed in a Cox's proportional hazard regression. The 5-year conditional RFS increased from 86% (95% confidence interval: 84-88) at diagnosis to 96% (95% confidence interval: 94-98) at 5 years after diagnosis. Women showed a 60% lower risk of recurrence than men and this effect was stable over time (P=0.39). Patients aged greater than or equal to 65 years had a 40% higher risk of recurrence than patients aged less than 65 years, and this effect was stable over time (P=0.65). Patients with tumour ulceration showed a 70% higher risk of recurrence than nonulcerated patients, but this effect disappeared after 2 years (P=0.04). For patients with T3-T4 CMM, the hazard ratios decreased over time and were similar to hazard ratio of patients with T2 CMM after 2 years and later. The decreasing impact of tumour thickness and ulceration over time could have important implications for CMM patients in terms of counselling and follow-up.
Collapse
Affiliation(s)
- Johan Lyth
- Research and Development Unit in Region Östergötland, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
757
|
Parlati L, Vallet-Pichard A, Batista R, Hernvann A, Sogni P, Pol S, Mallet V. Incidence of grade 3-4 liver injury under immune checkpoints inhibitors: A retrospective study. J Hepatol 2018; 69:1396-1397. [PMID: 30292476 DOI: 10.1016/j.jhep.2018.08.014] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 08/01/2018] [Accepted: 08/15/2018] [Indexed: 12/22/2022]
Affiliation(s)
- Lucia Parlati
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Assistance Publique - Hôpitaux de Paris (AP-HP), Hôpital Cochin, Hepatology Service, France
| | - Anais Vallet-Pichard
- Assistance Publique - Hôpitaux de Paris (AP-HP), Hôpital Cochin, Hepatology Service, France
| | - Rui Batista
- Assistance Publique - Hôpitaux de Paris (AP-HP), Hôpital Cochin, Pharmacy Service, Paris, France
| | - Alain Hernvann
- Assistance Publique - Hôpitaux de Paris (AP-HP), Hôpital Cochin, Biology Service, Paris, France
| | - Philippe Sogni
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Assistance Publique - Hôpitaux de Paris (AP-HP), Hôpital Cochin, Hepatology Service, France; Institut Pasteur, Inserm, Unité 1223, Paris, France
| | - Stanislas Pol
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Assistance Publique - Hôpitaux de Paris (AP-HP), Hôpital Cochin, Hepatology Service, France; Institut Pasteur, Inserm, Unité 1223, Paris, France
| | - Vincent Mallet
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Assistance Publique - Hôpitaux de Paris (AP-HP), Hôpital Cochin, Hepatology Service, France; Institut Pasteur, Inserm, Unité 1223, Paris, France.
| | | |
Collapse
|
758
|
van Koevering K, Prevedello DM, Carrau RL. Endoscopic endonasal approaches for the management of cranial base malignancies: histologically guided treatment and clinical outcomes. J Neurosurg Sci 2018; 62:667-681. [PMID: 29808637 DOI: 10.23736/s0390-5616.18.04507-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Malignancies of the skull base represent a highly diverse and challenging set of pathologies which exhibit a wide array of oncologic behavior. In recent decades, a number of important advances in treatment technique have evolved to improve oncologic outcomes and reduce morbidity in the treatment of these aggressive cancers. Intensity modulated radiation therapy has become the gold standard in radiotherapy owing to its precision planning and reduced morbidity. However heavy ion particle radiation (proton therapy, carbon ion, etc.) are recently emerging with promising results at the skull base related to the reduced exit dose to adjacent structures. Novel systemic therapeutics such as targeted and immunotherapies may dramatically alter the treatment paradigm for many of these pathologies. Nevertheless, most skull base malignancies remain surgical diseases. The evolution of the expanded endonasal approach for a minimally invasive surgical resection has proven validity in treating many of these pathologies when properly selected, and have largely supplanted open approaches owing to the reduced morbidity profile. In spite of these important advances, the most critical aspect in comprehensive treatment is a detailed understanding of the oncologic behavior and outcomes data for each of the specific histopathologies encountered at the skull base. The nuances in management strategy, histologic profile, and surveillance planning can be stratified through the development of a comprehensive, multidisciplinary skull base team to maximize therapeutic effect and minimize morbidity for each patient. This review aims to summarize the key body of data and approaches for each of the histologies frequently encountered in the skull base, while highlighting the value and technique of endonasal approaches.
Collapse
Affiliation(s)
- Kyle van Koevering
- Department of Otolaryngology, Head and Neck Surgery, Wexner Medical Center, Ohio State University, Columbus, OH, USA
| | - Daniel M Prevedello
- Department of Neurosurgery, Wexner Medical Center, Ohio State University, Columbus, OH, USA
| | - Ricardo L Carrau
- Department of Neurosurgery, Wexner Medical Center, Ohio State University, Columbus, OH, USA -
| |
Collapse
|
759
|
Grob JJ, Garbe C, Ascierto P, Larkin J, Dummer R, Schadendorf D. Adjuvant melanoma therapy with new drugs: should physicians continue to focus on metastatic disease or use it earlier in primary melanoma? Lancet Oncol 2018; 19:e720-e725. [DOI: 10.1016/s1470-2045(18)30596-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 08/01/2018] [Accepted: 08/02/2018] [Indexed: 02/06/2023]
|
760
|
Mackiewicz J, Burzykowski T, Iżycki D, Mackiewicz A. Re-induction using whole cell melanoma vaccine genetically modified to melanoma stem cells-like beyond recurrence extends long term survival of high risk resected patients - updated results. J Immunother Cancer 2018; 6:134. [PMID: 30486884 PMCID: PMC6264600 DOI: 10.1186/s40425-018-0456-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 11/16/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND AGI-101H is an allogeneic gene modified whole cell therapeutic melanoma vaccine, evaluated in over 400 melanoma patients in the adjuvant and therapeutic settings. We present updated long-term survival results from two single-arm, phase II adjuvant trials (Trial 3 and Trial 5) with the focus on treatment beyond recurrence of the disease. METHODS Patients with resected high-risk melanoma (stage IIIB-IV) were enrolled to Trial 3 (n = 99) and Trial 5 (n = 97). The primary endpoint was disease-free survival (DFS), and the secondary was overall survival (OS). In the induction phase, the vaccine was administered every 2 weeks (eight times), followed by the maintenance phase every month until progression. At progression, maintenance was continued or re-induction was applied with or without surgery. RESULTS In Trial 3, the 10-year DFS was equal to 33.0% overall and to 52.4, 25.0, and 8.7% for stage IIIB, IIIC, and stage IV patients, respectively. In Trial 5, the overall 10-year DFS was equal to 24.2%, and to 37.5, 18.0, and 17.6% for stage IIIB, IIIC, and stage IV patients, respectively. In Trial 3, the 10-year OS was equal to 42.3% overall, and to 59.5, 37.5, and 17.4% for stage IIIB, IIIC, and stage IV patients, respectively. In Trial 5, the 10-year OS was equal to 34.3% overall and to 46.9, 28.0, and 29.4% for stage IIIB, IIIC, and stage IV patients, respectively. Among the 65 patients of Trial 3 who developed progression, 43 received re-induction with (n = 22) or without (n = 21) surgery. Two patients received surgery without re-induction. All the 22 progressing patients, who did not receive re-induction, died. Among the 75 patients of Trial 5 who experienced progression, 39 received re-induction with (n = 21) or without (n = 18) surgery. Among the 36 progressing patients who did not receive the re-induction, 35 died. Surgery and re-induction reduced (independently) the increase of mortality after progression in both trials, with the effect of re-induction reaching statistical significance in Trial 5. CONCLUSIONS Vaccination beyond recurrence of the disease with additional re-induction combined with surgery or alone increased long term survival of melanoma patients. However, further studies on larger patient cohorts are required. TRIAL REGISTRATION Central Evidence of Clinical Trials (EudraCT Number 2008-003373-40 ).
Collapse
Affiliation(s)
- Jacek Mackiewicz
- Chair of Medical Biotechnology, University of Medical Sciences, 15 Garbary street, 61-866, Poznan, Poland. .,Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary street, 61-866, Poznan, Poland. .,Department of Medical and Experimental Oncology, Heliodor Świecicki University Hospital, Poznan University of Medical Sciences, Poland 15, 16/18 Grunwaldzka St, 60-780, Poznan, Poland. .,Department of Biology and Environmental Studies, University of Medical Sciences, 8 Rokietnicka street, 60-806, Poznan, Poland.
| | - Tomasz Burzykowski
- Interuniversity Institute for Biostatistics and statistical Bioinformatics, Hasselt University, 42 Martelarenlaan street, 3500, Diepenbeek, Belgium
| | - Dariusz Iżycki
- Chair of Medical Biotechnology, University of Medical Sciences, 15 Garbary street, 61-866, Poznan, Poland
| | - Andrzej Mackiewicz
- Chair of Medical Biotechnology, University of Medical Sciences, 15 Garbary street, 61-866, Poznan, Poland.,Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary street, 61-866, Poznan, Poland.,Department of Medical and Experimental Oncology, Heliodor Świecicki University Hospital, Poznan University of Medical Sciences, Poland 15, 16/18 Grunwaldzka St, 60-780, Poznan, Poland.,BioContract Sp z o.o., 36 Zambrowska street, 61-051, Poznan, Poland
| |
Collapse
|
761
|
Koelblinger P, Emberger M, Drach M, Cheng P, Lang R, Levesque M, Bauer J, Dummer R. Increased tumour cellPD‐L1 expression, macrophage and dendritic cell infiltration characterise the tumour microenvironment of ulcerated primary melanomas. J Eur Acad Dermatol Venereol 2018; 33:667-675. [DOI: 10.1111/jdv.15302] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 09/14/2018] [Indexed: 12/27/2022]
Affiliation(s)
- P. Koelblinger
- Department of Dermatology University Hospital of Zurich Zurich Switzerland
- Department of Dermatology Paracelsus Medical University Salzburg Austria
| | - M. Emberger
- Private Pathological Laboratory Salzburg Austria
| | - M. Drach
- Department of Dermatology University Hospital of Zurich Zurich Switzerland
| | - P.F. Cheng
- Department of Dermatology University Hospital of Zurich Zurich Switzerland
| | - R. Lang
- Department of Dermatology Paracelsus Medical University Salzburg Austria
| | - M.P. Levesque
- Department of Dermatology University Hospital of Zurich Zurich Switzerland
| | - J.W. Bauer
- Department of Dermatology Paracelsus Medical University Salzburg Austria
| | - R. Dummer
- Department of Dermatology University Hospital of Zurich Zurich Switzerland
| |
Collapse
|
762
|
Liu J, Rozeman EA, O'Donnell JS, Allen S, Fanchi L, Smyth MJ, Blank CU, Teng MWL. Batf3 + DCs and type I IFN are critical for the efficacy of neoadjuvant cancer immunotherapy. Oncoimmunology 2018; 8:e1546068. [PMID: 30713806 DOI: 10.1080/2162402x.2018.1546068] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 10/28/2018] [Accepted: 11/02/2018] [Indexed: 12/31/2022] Open
Abstract
New clinical trials are now evaluating the efficacy of neoadjuvant immunotherapy in the context of primary tumor surgery. Using the orthotopic 4T1.2 mouse model of spontaneously metastatic mammary cancer, we have shown that neoadjuvant immunotherapy and surgery was superior in the generation of tumor-specific CD8+ T cells and eradication of lethal metastases compared to surgery followed by adjuvant immunotherapy. However, the importance of host Batf3 and type I interferon (IFN) for long-term survival of mice following neoadjuvant immunotherapy is unknown. Here we demonstrated that loss of Batf3+ DCs or type I IFN receptor blockade in 4T1.2 tumor-bearing mice treated with neoadjuvant anti-PD-1+anti-CD137 immunotherapy reduced long-term survival with a corresponding reduction in tumor-specific CD8+ T cells producing effector cytokines in the primary tumor and in the periphery. Interestingly, we found all high-risk stage III melanoma patients relapsing after adjuvant or neoadjuvant ipilimumab+nivolumab within the OpACIN trial (NCT02437279) displayed low expression of Batf3+ DC-associated genes in pre-treatment tumor biopsies. Further focus should now be placed on validating the requirement of an intratumoral Batf3+ DC gene signature for response to neoadjuvant immunotherapy.
Collapse
Affiliation(s)
- Jing Liu
- Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Elisa A Rozeman
- Division of Molecular Oncology & Immunology, Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Jake S O'Donnell
- Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,School of Medicine, University of Queensland, Herston, Queensland, Australia
| | - Stacey Allen
- Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Lorenzo Fanchi
- Division of Molecular Oncology & Immunology, Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,School of Medicine, University of Queensland, Herston, Queensland, Australia
| | - Christian U Blank
- Division of Molecular Oncology & Immunology, Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Michele W L Teng
- Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,School of Medicine, University of Queensland, Herston, Queensland, Australia
| |
Collapse
|
763
|
Faut M, Jalving M, Diercks GF, Hospers GA, van Leeuwen BL, Been LB. Preoperative BRAF inhibition in patients with irresectable locally advanced stage III melanoma. Melanoma Manag 2018; 5:MMT08. [PMID: 30459939 PMCID: PMC6240848 DOI: 10.2217/mmt-2018-0002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 04/19/2018] [Indexed: 11/21/2022] Open
Abstract
Aim Neoadjuvant treatment of locally advanced disease with BRAF inhibitors is expected to increase the likelihood of a R0 resection. We present six patients with stage III unresectable melanoma, neoadjuvantly treated with BRAF inhibitors. Methods Patients with unresectable, BRAF-mutated, stage III melanoma, were treated with BRAF inhibitors between 2012 and 2015. Unresectability was determined based on clinical and/or radiological findings. At maximal response, resection was performed. The specimen was reviewed to determine the degree of response. Results In five of six patients a radical resection was achieved. Postoperative complications were unremarkable. In five of six resected specimens, vital tumor tissue was found. Conclusion Neoadjuvant BRAF inhibitor treatment of locally advanced melanoma is feasible and has the potential to facilitate an R0 resection.
Collapse
Affiliation(s)
- Marloes Faut
- Department of Surgical Oncology, Department of Surgery, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30.001, 9700RB Groningen, The Netherlands.,Department of Surgical Oncology, Department of Surgery, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30.001, 9700RB Groningen, The Netherlands
| | - Mathilde Jalving
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30.001, 9700RB Groningen, The Netherlands.,Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30.001, 9700RB Groningen, The Netherlands
| | - Gilles F Diercks
- Department of Pathology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30.001, 9700RB Groningen, The Netherlands.,Department of Pathology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30.001, 9700RB Groningen, The Netherlands
| | - Geke A Hospers
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30.001, 9700RB Groningen, The Netherlands.,Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30.001, 9700RB Groningen, The Netherlands
| | - Barbara L van Leeuwen
- Department of Surgical Oncology, Department of Surgery, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30.001, 9700RB Groningen, The Netherlands.,Department of Surgical Oncology, Department of Surgery, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30.001, 9700RB Groningen, The Netherlands
| | - Lukas B Been
- Department of Surgical Oncology, Department of Surgery, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30.001, 9700RB Groningen, The Netherlands.,Department of Surgical Oncology, Department of Surgery, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30.001, 9700RB Groningen, The Netherlands
| |
Collapse
|
764
|
Health-related quality of life analysis in stage III melanoma patients treated with adjuvant dendritic cell therapy. Clin Transl Oncol 2018; 21:774-780. [PMID: 30465182 DOI: 10.1007/s12094-018-1987-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 11/09/2018] [Indexed: 10/27/2022]
Abstract
BACKGROUND Health-related quality of life (HRQoL) is an important issue in the rapidly evolving field of adjuvant treatment for stage III melanoma. Dendritic cell vaccination is one of the adjuvant forms of therapy currently investigated. METHODS We enrolled adults with stage III melanoma to receive adjuvant dendritic cell vaccination after a complete radical lymph node dissection. HRQoL assessment was one of the secondary endpoints of this trial and investigated with the EORTC-QLQ-C30 questionnaire at baseline and week 26. RESULTS Fifteen patients with a median age of 50 years were included in the study, with twelve evaluable patients on study at time of the second questionnaire. Global health status and role functioning improved clinically relevant with a mean difference of 15 (p = 0.010) and 26 points (p = 0.005), respectively. DISCUSSION Despite the small number of patients, we found a clinically relevant improved global health status. Besides, compared to the other investigated therapies, toxicity of dendritic cell vaccination is low, which supports our finding. CONCLUSION This is the first description of HRQoL in melanoma patients receiving dendritic cell vaccination. We show the expected improvement in global health status after surgical treatment of stage III melanoma. Thus, adjuvant dendritic cell vaccination does not seem to hamper this improvement, as shown in our small explorative study.
Collapse
|
765
|
Izzedine H, Mathian A, Champiat S, Picard C, Mateus C, Routier E, Varga A, Malka D, Leary A, Michels J, Michot JM, Marabelle A, Lambotte O, Amoura Z, Soria JC, Kaaki S, Quellard N, Goujon JM, Brocheriou I. Renal toxicities associated with pembrolizumab. Clin Kidney J 2018; 12:81-88. [PMID: 30746132 PMCID: PMC6366307 DOI: 10.1093/ckj/sfy100] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 09/05/2018] [Indexed: 02/07/2023] Open
Abstract
Objective Expanded clinical experience with patients treated by pembrolizumab has accumulated. However, renal toxicities associated with this anti-programmed cell death 1 agent are poorly described because kidney histology is rarely sought. As a nephrology referral centre, we aimed to describe the clinic-biological and histopathological characteristics of pembrolizumab-related nephropathy and its response to treatment. Methods We conducted a monocentric large case series study, including all pembrolizumab-treated cancer patients presenting a renal toxicity addressed to our centre from 2015 to 2017. Results A total of 12 patients (7 men) out of 676 pembrolizumab-treated patients (incidence 1.77%) were included (median age 69.75 years). Patients were referred for acute kidney injury (n = 10) and/or proteinuria (n = 2). A kidney biopsy was performed in all patients, with a median duration of use of 9 months (range 1–24 months) after the beginning of treatment. Biopsy showed that four patients had acute interstitial nephritis (AIN), whereas five had acute tubular injury (ATI) alone, one had minimal change disease (MCD) and ATI, and one had MCD alone. Pembrolizumab withdrawal coupled with corticosteroid therapy was the most effective treatment for kidney function recovery. Drug reintroduction resulted in a more severe recurrence of AIN in one patient who required maintenance of pembrolizumab. Two patients died of cancer progression with one of them developing severe renal failure requiring dialysis. Conclusion In our series, ATI, AIN and MCD are the most frequent forms of kidney involvement under pembrolizumab therapy. Kidney dysfunction is usually isolated but can be severe. Use of corticosteroids in case of AIN improves the glomerular filtration rate.
Collapse
Affiliation(s)
- Hassan Izzedine
- Department of Nephrology, Peupliers Private Hospital, Ramsay Générale de Santé, Paris, France.,UPEC (Université Paris Est Créteil), INSERM U955, Institut Mondor de Recherche Biomdicale (IMRB), Equipe, Créteil, France
| | - Alexis Mathian
- Department of Internal Medicine, Pitie-Salpetriere Hospital, Paris, France
| | - Stephane Champiat
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France
| | - Cécile Picard
- Department of Pathology, Pitie-Salpetriere Hospital, Paris, France
| | - Christine Mateus
- Department of Supportive Care, Gustave Roussy, Villejuif, France
| | - Emilie Routier
- Department of Medical Oncology, Dermatology Unit, Gustave Roussy, Villejuif, France
| | - Andrea Varga
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France
| | - David Malka
- Department of Medical Oncology, Gastrointestinal Cancer Group, Gustave Roussy, Villejuif, France
| | - Alexandra Leary
- Department of Medical Oncology, Gynecology Unit, Gustave Roussy, Villejuif, France
| | - Judith Michels
- Department of Medical Oncology, Gynecology Unit, Gustave Roussy, Villejuif, France
| | - Jean-Marie Michot
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France
| | | | - Olivier Lambotte
- Department of Internal Medicine and Clinical Immunology, Bicetre University Hospital, Le Kremlin Bicêtre, France
| | - Zahir Amoura
- Department of Internal Medicine, Pitie-Salpetriere Hospital, Paris, France
| | | | - Sihem Kaaki
- Departement of Pathology, Electron Microscopy Unit, CHU Poitiers, Poitiers, France
| | - Nathalie Quellard
- Departement of Pathology, Electron Microscopy Unit, CHU Poitiers, Poitiers, France
| | - Jean-Michel Goujon
- Departement of Pathology, Electron Microscopy Unit, CHU Poitiers, Poitiers, France
| | | |
Collapse
|
766
|
Kudchadkar RR, Michielin O, van Akkooi ACJ. Practice-Changing Developments in Stage III Melanoma: Surgery, Adjuvant Targeted Therapy, and Immunotherapy. Am Soc Clin Oncol Educ Book 2018; 38:759-762. [PMID: 30231370 DOI: 10.1200/edbk_200241] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In this article, we will focus on the practice-changing developments for stage III melanoma, from the use of the sentinel node (SN) biopsy to complete lymph node dissection (CLND) and upcoming adjuvant therapies. MSLT-1 (Multicenter Selective Lymphadenectomy Trial-1) was the first and only prospective randomized controlled trial to examine whether the SN biopsy has any notable melanoma-specific survival benefit (primary endpoint). MSLT-1 randomly assigned 2,001 patients to undergo either wide local excision (WLE) and an SN biopsy or WLE and nodal observation. Two prospective randomized controlled trials have examined the potential benefit for immediate CLND versus delayed CLND after sequential observation. Both the DECOG-SLT and MSLT-2 trials failed to demonstrate a notable benefit for immediate CLND; therefore, sequential follow-up with ultrasonography and a delayed CLND in the case of relapse should be considered the new standard of care. The CheckMate 238 study demonstrated a notable benefit for adjuvant nivolumab in terms of 18-month relapse-free survival (RFS) rates compared with high-dose adjuvant ipilimumab. Single-agent adjuvant BRAF inhibition has been examined and failed to improve RFS. However, the COMBI-AD study did demonstrate a substantial benefit for combination BRAF and MEK inhibition for patients with BRAF-mutated resected stage IIIA to IIIC melanoma.
Collapse
Affiliation(s)
- Ragini R Kudchadkar
- From the Department of Hematology/Oncology, Winship Cancer Institute, Emory University, Atlanta, GA; Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland; Department of Surgical Oncology, Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, Netherlands
| | - Olivier Michielin
- From the Department of Hematology/Oncology, Winship Cancer Institute, Emory University, Atlanta, GA; Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland; Department of Surgical Oncology, Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, Netherlands
| | - Alexander C J van Akkooi
- From the Department of Hematology/Oncology, Winship Cancer Institute, Emory University, Atlanta, GA; Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland; Department of Surgical Oncology, Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, Netherlands
| |
Collapse
|
767
|
Amaria RN, Reddy SM, Tawbi HA, Davies MA, Ross MI, Glitza IC, Cormier JN, Lewis C, Hwu WJ, Hanna E, Diab A, Wong MK, Royal R, Gross N, Weber R, Lai SY, Ehlers R, Blando J, Milton DR, Woodman S, Kageyama R, Wells DK, Hwu P, Patel SP, Lucci A, Hessel A, Lee JE, Gershenwald J, Simpson L, Burton EM, Posada L, Haydu L, Wang L, Zhang S, Lazar AJ, Hudgens CW, Gopalakrishnan V, Reuben A, Andrews MC, Spencer CN, Prieto V, Sharma P, Allison J, Tetzlaff MT, Wargo JA. Neoadjuvant immune checkpoint blockade in high-risk resectable melanoma. Nat Med 2018; 24:1649-1654. [PMID: 30297909 PMCID: PMC6481682 DOI: 10.1038/s41591-018-0197-1] [Citation(s) in RCA: 609] [Impact Index Per Article: 87.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 08/21/2018] [Indexed: 12/17/2022]
Abstract
Preclinical studies suggest that treatment with neoadjuvant immune checkpoint blockade is associated with enhanced survival and antigen-specific T cell responses compared with adjuvant treatment1; however, optimal regimens have not been defined. Here we report results from a randomized phase 2 study of neoadjuvant nivolumab versus combined ipilimumab with nivolumab in 23 patients with high-risk resectable melanoma ( NCT02519322 ). RECIST overall response rates (ORR), pathologic complete response rates (pCR), treatment-related adverse events (trAEs) and immune correlates of response were assessed. Treatment with combined ipilimumab and nivolumab yielded high response rates (RECIST ORR 73%, pCR 45%) but substantial toxicity (73% grade 3 trAEs), whereas treatment with nivolumab monotherapy yielded modest responses (ORR 25%, pCR 25%) and low toxicity (8% grade 3 trAEs). Immune correlates of response were identified, demonstrating higher lymphoid infiltrates in responders to both therapies and a more clonal and diverse T cell infiltrate in responders to nivolumab monotherapy. These results describe the feasibility of neoadjuvant immune checkpoint blockade in melanoma and emphasize the need for additional studies to optimize treatment regimens and to validate putative biomarkers.
Collapse
Affiliation(s)
- Rodabe N. Amaria
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Sangeetha M. Reddy
- Department of Breast Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Hussein A. Tawbi
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Michael A. Davies
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Merrick I. Ross
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Isabella C. Glitza
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Janice N. Cormier
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Carol Lewis
- Department of Head and Neck Surgery, MD Anderson Cancer Center, Houston, Texas, USA
| | - Wen-Jen Hwu
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Ehab Hanna
- Department of Head and Neck Surgery, MD Anderson Cancer Center, Houston, Texas, USA
| | - Adi Diab
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Michael K. Wong
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Richard Royal
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Neil Gross
- Department of Head and Neck Surgery, MD Anderson Cancer Center, Houston, Texas, USA
| | - Randal Weber
- Department of Head and Neck Surgery, MD Anderson Cancer Center, Houston, Texas, USA
| | - Stephen Y. Lai
- Department of Head and Neck Surgery, MD Anderson Cancer Center, Houston, Texas, USA
| | - Richard Ehlers
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Jorge Blando
- Department of Immunology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Denái R. Milton
- Department of Biostatistics, MD Anderson Cancer Center, Houston, Texas, USA
| | - Scott Woodman
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Robin Kageyama
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | - Danny K. Wells
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | - Patrick Hwu
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Sapna P. Patel
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Anthony Lucci
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Amy Hessel
- Department of Head and Neck Surgery, MD Anderson Cancer Center, Houston, Texas, USA
| | - Jeffrey E. Lee
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Jeffrey Gershenwald
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Lauren Simpson
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Elizabeth M. Burton
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Liberty Posada
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Lauren Haydu
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Linghua Wang
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, Texas, USA
| | - Shaojun Zhang
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | - Alexandre Reuben
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Miles C. Andrews
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Victor Prieto
- Department of Immunology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Padmanee Sharma
- Department of Immunology, MD Anderson Cancer Center, Houston, Texas, USA
- Department of Genitourinary Cancers, MD Anderson Cancer Center, Houston, Texas, USA
| | - James Allison
- Department of Immunology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Michael T. Tetzlaff
- Department of Pathology, MD Anderson Cancer Center, Houston, Texas, USA
- Department of Translational and Molecular Pathology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Jennifer A. Wargo
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
768
|
Ziogas DE, Kyrochristos ID, Roukos DH. Discovering novel valid biomarkers and drugs in patient-centric genomic trials: the new epoch of precision surgical oncology. Drug Discov Today 2018; 23:1848-1872. [PMID: 30077778 DOI: 10.1016/j.drudis.2018.07.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 07/10/2018] [Accepted: 07/26/2018] [Indexed: 12/16/2022]
Abstract
Despite standardization of multimodal treatment and approval of several targeted drugs for resectable, non-metastatic cancer (M0 patients), intrinsic and acquired resistance and relapse rates remain high, even in early-stage aggressive tumors. Genome analysis could overcome these unmet needs. Our comprehensive review underlines the controversy on stable or spatiotemporally evolving clones as well as promising yet inconclusive data on genome-based biomarkers and drug development. We propose clinicogenomic trials in M0 patients for the validation of intratumor heterogeneity (ITH), circulating genomic subclones (cGSs) and intra-patient genomic heterogeneity (IPGH) as biomarkers and simultaneous discovery of novel oncotargets. This evidence-based strategy highlights the coming of precision surgical oncology with a future perspective of understanding and disrupting deregulated transcriptional networks.
Collapse
Affiliation(s)
- Demosthenes E Ziogas
- Centre for Biosystems and Genome Network Medicine, Ioannina University, Ioannina, Greece; Department of Surgery, 'G. Hatzikosta' General Hospital, Ioannina, Greece
| | - Ioannis D Kyrochristos
- Centre for Biosystems and Genome Network Medicine, Ioannina University, Ioannina, Greece; Department of Surgery, Ioannina University Hospital, Ioannina, Greece
| | - Dimitrios H Roukos
- Centre for Biosystems and Genome Network Medicine, Ioannina University, Ioannina, Greece; Department of Surgery, Ioannina University Hospital, Ioannina, Greece; Department of Systems Biology, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece.
| |
Collapse
|
769
|
Swetter SM, Tsao H, Bichakjian CK, Curiel-Lewandrowski C, Elder DE, Gershenwald JE, Guild V, Grant-Kels JM, Halpern AC, Johnson TM, Sober AJ, Thompson JA, Wisco OJ, Wyatt S, Hu S, Lamina T. Guidelines of care for the management of primary cutaneous melanoma. J Am Acad Dermatol 2018; 80:208-250. [PMID: 30392755 DOI: 10.1016/j.jaad.2018.08.055] [Citation(s) in RCA: 394] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 08/28/2018] [Accepted: 08/29/2018] [Indexed: 12/12/2022]
Abstract
The incidence of primary cutaneous melanoma continues to increase each year. Melanoma accounts for the majority of skin cancer-related deaths, but treatment is usually curative following early detection of disease. In this American Academy of Dermatology clinical practice guideline, updated treatment recommendations are provided for patients with primary cutaneous melanoma (American Joint Committee on Cancer stages 0-IIC and pathologic stage III by virtue of a positive sentinel lymph node biopsy). Biopsy techniques for a lesion that is clinically suggestive of melanoma are reviewed, as are recommendations for the histopathologic interpretation of cutaneous melanoma. The use of laboratory, molecular, and imaging tests is examined in the initial work-up of patients with newly diagnosed melanoma and for follow-up of asymptomatic patients. With regard to treatment of primary cutaneous melanoma, recommendations for surgical margins and the concepts of staged excision (including Mohs micrographic surgery) and nonsurgical treatments for melanoma in situ, lentigo maligna type (including topical imiquimod and radiation therapy), are updated. The role of sentinel lymph node biopsy as a staging technique for cutaneous melanoma is described, with recommendations for its use in clinical practice. Finally, current data regarding pregnancy and melanoma, genetic testing for familial melanoma, and management of dermatologic toxicities related to novel targeted agents and immunotherapies for patients with advanced disease are summarized.
Collapse
Affiliation(s)
- Susan M Swetter
- Department of Dermatology, Stanford University Medical Center and Cancer Institute, Stanford, California; Veterans Affairs Palo Alto Health Care System, Palo Alto, California.
| | - Hensin Tsao
- Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts; Wellman Center for Photomedicine, Boston, Massachusetts
| | - Christopher K Bichakjian
- Department of Dermatology, University of Michigan Health System, Ann Arbor, Michigan; Comprehensive Cancer Center, Ann Arbor, Michigan
| | - Clara Curiel-Lewandrowski
- Division of Dermatology, University of Arizona, Tucson, Arizona; University of Arizona Cancer Center, Tucson, Arizona
| | - David E Elder
- Department of Dermatology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania; Department of Pathology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jeffrey E Gershenwald
- Department of Surgical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas; Department of Cancer Biology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | | | - Jane M Grant-Kels
- Department of Dermatology, University of Connecticut Health Center, Farmington, Connecticut; Department of Pathology, University of Connecticut Health Center, Farmington, Connecticut; Department of Pediatrics, University of Connecticut Health Center, Farmington, Connecticut
| | - Allan C Halpern
- Department of Dermatology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Timothy M Johnson
- Department of Dermatology, University of Michigan Health System, Ann Arbor, Michigan; Comprehensive Cancer Center, Ann Arbor, Michigan
| | - Arthur J Sober
- Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - John A Thompson
- Division of Oncology, University of Washington, Seattle, Washington; Seattle Cancer Care Alliance, Seattle, Washington
| | - Oliver J Wisco
- Department of Dermatology, Oregon Health and Science University, Portland, Oregon
| | | | - Shasa Hu
- Department of Dermatology, University of Miami Health System, Miami, Florida
| | - Toyin Lamina
- American Academy of Dermatology, Rosemont, Illinois
| |
Collapse
|
770
|
Soularue E, Lepage P, Colombel JF, Coutzac C, Faleck D, Marthey L, Collins M, Chaput N, Robert C, Carbonnel F. Enterocolitis due to immune checkpoint inhibitors: a systematic review. Gut 2018; 67:2056-2067. [PMID: 30131322 DOI: 10.1136/gutjnl-2018-316948] [Citation(s) in RCA: 178] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 07/11/2018] [Accepted: 07/19/2018] [Indexed: 12/14/2022]
Abstract
Immune checkpoint inhibitors targeting cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) and programmed death-1 (PD-1)/ligand are increasingly used to treat several types of cancer. These drugs enhance antitumour T-cell activity and therefore induce immune-related adverse effects (irAE), of which gastrointestinal (GI) irAE are among the most frequent and severe. This systematic literature review summarises the clinical manifestations, management and pathophysiology of GI irAE due to immune checkpoint inhibitors. GI irAE induced by anti-CTLA-4 are frequent, potentially severe and resemble IBD, whereas those induced by PD-1 blockade seem to be less frequent and clinically more diverse. Baseline symbiotic gut microbiota is associated with an enhanced antitumour response to immune checkpoint inhibitors and an increased susceptibility to developing enterocolitis, in patients treated with anti-CTLA-4. These findings open new perspectives for possible manipulation of the gut microbiota in order to better identify responders to immune checkpoint inhibitors and to increase their efficacy and safety.
Collapse
Affiliation(s)
- Emilie Soularue
- Department of Gastroenterology, Kremlin Bicêtre Hospital, Assistance Publique- Hôpitaux de Paris (AP-HP), Le Kremlin Bicêtre, France.,Faculté de Médecine, University Paris-Saclay, Le Kremlin Bicêtre, France
| | - Patricia Lepage
- Micalis Institute, INRA, AgroParisTech, University Paris-Saclay, Jouy-en- Josas, France
| | - Jean Frederic Colombel
- Helmsley Inflammatory Bowel Disease Center, Icahn Medical School of Medicine at Mount Sinai, New York, USA
| | - Clelia Coutzac
- Laboratory of Immunomonitoring in Oncology and CNRS-UMS 3655 and INSERM-US23, Villejuif, France
| | - David Faleck
- Helmsley Inflammatory Bowel Disease Center, Icahn Medical School of Medicine at Mount Sinai, New York, USA
| | - Lysiane Marthey
- Department of Gastroenterology, Kremlin Bicêtre Hospital, Assistance Publique- Hôpitaux de Paris (AP-HP), Le Kremlin Bicêtre, France
| | - Michael Collins
- Department of Gastroenterology, Kremlin Bicêtre Hospital, Assistance Publique- Hôpitaux de Paris (AP-HP), Le Kremlin Bicêtre, France.,Faculté de Médecine, University Paris-Saclay, Le Kremlin Bicêtre, France
| | - Nathalie Chaput
- Laboratory of Immunomonitoring in Oncology and CNRS-UMS 3655 and INSERM-US23, Villejuif, France.,Faculté de Pharmacie, University Paris-Saclay, Chatenay-Malabry, France
| | - Caroline Robert
- Faculté de Médecine, University Paris-Saclay, Le Kremlin Bicêtre, France.,Départment of Medecine, Dermatology Unit, Villejuif, France
| | - Franck Carbonnel
- Department of Gastroenterology, Kremlin Bicêtre Hospital, Assistance Publique- Hôpitaux de Paris (AP-HP), Le Kremlin Bicêtre, France.,Faculté de Médecine, University Paris-Saclay, Le Kremlin Bicêtre, France
| |
Collapse
|
771
|
Solomon BL, Garrido-Laguna I. TIGIT: a novel immunotherapy target moving from bench to bedside. Cancer Immunol Immunother 2018; 67:1659-1667. [PMID: 30232519 PMCID: PMC11028339 DOI: 10.1007/s00262-018-2246-5] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 09/12/2018] [Indexed: 12/28/2022]
Abstract
Treatment strategies for patients with advanced solid tumors have traditionally been based on three different paradigms: surgery, cytotoxics (chemotherapy or radiation therapy) and targeted therapies. Immunotherapy has emerged as a novel treatment paradigm in our armamentarium. Unfortunately, most patients still do not benefit from immunotherapy. These patients often have "cold tumors" characterized by a paucity of effector T cells in the tumor microenvironment, low mutational load, low neoantigen burden and often an immunosuppressive tumor microenvironment. TIGIT is an immunoreceptor inhibitory checkpoint that has been implicated in tumor immunosurveillance. Expression of TIGIT has been demonstrated in both NK cells and T cells and plays a role in their activation and maturation. TIGIT competes with immunoactivator receptor CD226 (DNAM-1) for the same set of ligands: CD155 (PVR or poliovirus receptor) and CD112 (Nectin-2 or PVRL2). TIGIT's role in tumor immunosurveillance is analogous to the PD-1/PD-L1 axis in tumor immunosuppression. Both TIGIT and PD-1 are upregulated in a variety of different cancers. Anti-TIGIT antibodies have demonstrated synergy with anti-PD-1/PD-L1 antibodies in pre-clinical models. Currently, there are multiple first-in-man phase I trials hoping to exploit this new pathway and improve response rates with existing immunotherapies.
Collapse
Affiliation(s)
- Benjamin L Solomon
- Oncology Division, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah School of Medicine, 2000 Circle of Hope, Suite 2100, Salt Lake City, UT, 84112, USA.
| | - Ignacio Garrido-Laguna
- Oncology Division, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah School of Medicine, 2000 Circle of Hope, Suite 2100, Salt Lake City, UT, 84112, USA
- Center for Investigational Therapeutics, Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| |
Collapse
|
772
|
Musoro JZ, Bottomley A, Coens C, Eggermont AMM, King MT, Cocks K, Sprangers MAG, Groenvold M, Velikova G, Flechtner HH, Brandberg Y. Interpreting European Organisation for Research and Treatment for Cancer Quality of life Questionnaire core 30 scores as minimally importantly different for patients with malignant melanoma. Eur J Cancer 2018; 104:169-181. [DOI: 10.1016/j.ejca.2018.09.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 08/14/2018] [Accepted: 09/10/2018] [Indexed: 12/18/2022]
|
773
|
McKean MA, Amaria RN. Multidisciplinary treatment strategies in high-risk resectable melanoma: Role of adjuvant and neoadjuvant therapy. Cancer Treat Rev 2018; 70:144-153. [DOI: 10.1016/j.ctrv.2018.08.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 08/27/2018] [Accepted: 08/28/2018] [Indexed: 12/17/2022]
|
774
|
Gonzalez A. Sentinel Lymph Node Biopsy: Past and Present Implications for the Management of Cutaneous Melanoma with Nodal Metastasis. Am J Clin Dermatol 2018; 19:24-30. [PMID: 30374897 PMCID: PMC6244615 DOI: 10.1007/s40257-018-0379-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Although significant progress has been made in the understanding of melanoma pathophysiology and therapy, patients with metastatic melanoma still have a poor prognosis. The management of regional nodes remains a matter of debate. By replacing elective lymph node dissection, sentinel lymph node biopsy has revolutionized the treatment of malignant melanoma. In this paper, the history of the procedure is traced, and the indication for completion lymphadenectomy after positive sentinel node biopsy is discussed in light of the recent studies that addressed this issue. The role of adjuvant therapies in the management of patients with stage III melanoma is also discussed.
Collapse
Affiliation(s)
- Abel Gonzalez
- Mohs Surgery Unit, Institute of Oncology Angel H. Roffo, University of Buenos Aires, Av San Martin 5481, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina.
- Unit of Cutaneous Oncology and Mohs Surgery, Institute Alexander Fleming, Conesa 1003, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
775
|
Faut M, Kruijff S, Hoekstra HJ, van Ginkel RJ, Been LB, van Leeuwen BL. Pelvic lymph node dissection in metastatic melanoma to the groin should not be abandoned yet. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2018; 44:1779-1785. [PMID: 30054111 DOI: 10.1016/j.ejso.2018.06.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 03/02/2018] [Accepted: 06/27/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND In recent years there has been a plea to abandon the pelvic lymph node dissection in the treatment of patients with metastatic melanoma to the groin. A trend towards a conservative surgical treatment is already evolving in several European countries. The purpose of this study is to identify factors associated with pelvic nodal involvement, in order to improve selection of patients whom might benefit from a pelvic nodal dissection. METHODS A retrospective analysis was performed on prospectively collected data concerning patients who underwent an inguinal lymph node dissection (ILND) with pelvic lymph node dissection for metastatic melanoma at the University Medical Center Groningen. Multivariable logistic regression analysis was performed to determine factors associated with pelvic nodal involvement. Diagnostic accuracy was calculated for 18F-FDG PET + contrast enhanced CT-scan and 18F-FDG PET + low dose CT-scan. RESULTS Two-hundred-and-twenty-six ILND's were performed in 223 patients. The most common histologic subtype was superficial spreading melanoma (42.6%). In patients with micrometastatic disease, 15.7% had pelvic nodal involvement vs 28.2% in patients with macrometastatic disease (p: 0.030). None of the characteristics known prior to the ILND, were associated with pelvic nodal involvement. Imaging methods were unable to accurately predict pelvic nodal involvement. Negative predictive value was 78% for 18F-FDG PET + low dose CT-scan and 86% for an 18F-FDG PET + contrast enhanced CT-scan. CONCLUSION There are no patient- or tumor characteristics available that can predict pelvic nodal involvement in patients with melanoma metastasis to the groin. As no imaging technique is able to predict pelvic nodal involvement it seems unjust to abandon the pelvic lymph node dissection.
Collapse
Affiliation(s)
- M Faut
- Department of Surgical Oncology, Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - S Kruijff
- Department of Surgical Oncology, Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - H J Hoekstra
- Department of Surgical Oncology, Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - R J van Ginkel
- Department of Surgical Oncology, Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - L B Been
- Department of Surgical Oncology, Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - B L van Leeuwen
- Department of Surgical Oncology, Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
776
|
Wang DY, Mooradian MJ, Kim D, Shah NJ, Fenton SE, Conry RM, Mehta R, Silk AW, Zhou A, Compton ML, Al-Rohil RN, Lee S, Voorhees AL, Ha L, McKee S, Norrell JT, Mehnert J, Puzanov I, Sosman JA, Chandra S, Gibney GT, Rapisuwon S, Eroglu Z, Sullivan R, Johnson DB. Clinical characterization of colitis arising from anti-PD-1 based therapy. Oncoimmunology 2018; 8:e1524695. [PMID: 30546965 PMCID: PMC6287774 DOI: 10.1080/2162402x.2018.1524695] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 09/10/2018] [Accepted: 09/12/2018] [Indexed: 12/17/2022] Open
Abstract
Colitis is a frequent, clinically-significant immune-related adverse event caused by anti-programmed death-1 (PD-1). The clinical features, timing, and management of colitis with anti-PD-1-based regimens are not well-characterized. Patients with advanced melanoma that received either anti-PD-1 monotherapy ("monotherapy") or combined with ipilimumab ("combination therapy") were screened from 8 academic medical centers, to identify those with clinically-relevant colitis (colitis requiring systemic steroids). Of 1261 patients who received anti-PD-1-based therapy, 109 experienced colitis. The incidence was 3.2% (30/937) and 24.4% (79/324) in the monotherapy and combination therapy cohorts, respectively. Patients with colitis from combination therapy had significantly earlier symptom onset (7.2 weeks vs 25.4 weeks, p < 0.0001), received higher steroid doses (median prednisone equivalent 1.5 mg/kg vs 1.0 mg/kg, p = 0.0015) and experienced longer steroid tapers (median 6.0 vs 4.0 weeks, p = 0.0065) compared to monotherapy. Infliximab use and steroid-dose escalation occurred more frequently in the combination therapy cohort compared to monotherapy. Nearly all patients had resolution of their symptoms although one patient died from complications. Anti-PD-1 associated colitis has a variable clinical presentation, and is more frequent and severe when associated with combination therapy. This variability in checkpoint-inhibitor associated colitis suggests that further optimization of treatment algorithms is needed.
Collapse
Affiliation(s)
- Daniel Y Wang
- Department of Medicine, Vanderbilt University, Nashville, TN, USA,CONTACT Douglas B. Johnson Vanderbilt University Medical Center, 2220 Pierce Ave, 777 Preston Research Bldg. Nashville TN 37232
| | | | - DaeWon Kim
- Department of Medicine, Moffitt Cancer Center, Tampa, FL, USA
| | - Neil J Shah
- Department of Medicine, Georgetown University-Lombardi Cancer Center, Washington, DC, USA
| | - Sarah E Fenton
- Department of Medicine, Northwestern University, Chicago, IL, USA
| | - Robert M Conry
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rutika Mehta
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, IL, USA
| | - Ann W. Silk
- Department of Medicine, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Alice Zhou
- Department of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Margaret L Compton
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN, USA
| | - Rami N Al-Rohil
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN, USA
| | - Sunyoung Lee
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, IL, USA
| | | | - Lisa Ha
- Department of Medicine, Northwestern University, Chicago, IL, USA
| | - Svetlana McKee
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jacqueline T Norrell
- Department of Medicine, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Janice Mehnert
- Department of Medicine, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Igor Puzanov
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, IL, USA
| | - Jeffrey A Sosman
- Department of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Geoffrey T Gibney
- Department of Medicine, Georgetown University-Lombardi Cancer Center, Washington, DC, USA
| | - Suthee Rapisuwon
- Department of Medicine, Georgetown University-Lombardi Cancer Center, Washington, DC, USA
| | - Zeynep Eroglu
- Department of Medicine, Moffitt Cancer Center, Tampa, FL, USA
| | - Ryan Sullivan
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | | |
Collapse
|
777
|
Middleton MR. Long-term survival with anti-PD-1-based immunotherapy, but what is the best approach? Lancet Oncol 2018; 19:1424-1426. [PMID: 30361171 DOI: 10.1016/s1470-2045(18)30753-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 10/05/2018] [Indexed: 11/25/2022]
Affiliation(s)
- Mark R Middleton
- Department of Oncology, University of Oxford, Old Road Campus, Oxford, OX3 9DU, UK.
| |
Collapse
|
778
|
Frerich B. [Treatment of cutaneous malignant melanoma in the head and neck region : An update]. HNO 2018; 66:857-873. [PMID: 30302496 DOI: 10.1007/s00106-018-0573-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Major progress has been made in the treatment of malignant melanoma during recent years. On the one hand, reliable evidence-based recommendations regarding surgical resection as well as lymph node management have been established on the basis of results from randomized multicenter studies. On the other hand, the advent of targeted therapies has led to spectacular improvements in the treatment of metastasized melanomas. Knowledge concerning treatment algorithms and updates thereof is also essential for maxillofacial surgeons. The German S3 guideline on diagnosis and treatment of melanoma was published in 2013 and revised in 2016 and 2018. This article informs readers about these developments and addresses the items relevant for treatment of head and neck melanomas in particular.
Collapse
Affiliation(s)
- B Frerich
- Klinik und Poliklinik für Mund‑, Kiefer- und Plastische Gesichtschirurgie, Universitätsmedizin Rostock, Schillingallee 35, 18057, Rostock, Deutschland.
| |
Collapse
|
779
|
Verver D, van Klaveren D, Franke V, van Akkooi ACJ, Rutkowski P, Keilholz U, Eggermont AMM, Nijsten T, Grünhagen DJ, Verhoef C. Development and validation of a nomogram to predict recurrence and melanoma-specific mortality in patients with negative sentinel lymph nodes. Br J Surg 2018; 106:217-225. [PMID: 30307046 PMCID: PMC6585628 DOI: 10.1002/bjs.10995] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/04/2018] [Accepted: 08/08/2018] [Indexed: 12/21/2022]
Abstract
Background Patients with melanoma and negative sentinel nodes (SNs) have varying outcomes, dependent on several prognostic factors. Considering all these factors in a prediction model might aid in identifying patients who could benefit from a personalized treatment strategy. The objective was to construct and validate a nomogram for recurrence and melanoma‐specific mortality (MSM) in patients with melanoma and negative SNs. Methods A total of 3220 patients with negative SNs were identified from a cohort of 4124 patients from four EORTC Melanoma Group centres who underwent sentinel lymph node biopsy. Prognostic factors for recurrence and MSM were studied with Cox regression analysis. Significant factors were incorporated in the models. Performance was assessed by discrimination (c‐index) and calibration in cross‐validation across the four centres. A nomogram was developed for graphical presentation. Results There were 3180 eligible patients. The final prediction model for recurrence and the calibrated model for MSM included three independent prognostic factors: ulceration, anatomical location and Breslow thickness. The c‐index was 0·74 for recurrence and 0·76 for the calibrated MSM model. Cross‐validation across the four centres showed reasonable model performance. A nomogram was developed based on these models. One‐third of the patients had a 5‐year recurrence probability of 8·2 per cent or less, and one‐third had a recurrence probability of 23·0 per cent or more. Conclusion A nomogram for predicting recurrence and MSM in patients with melanoma and negative SNs was constructed and validated. It could provide personalized estimates useful for tailoring surveillance strategies (reduce or increase intensity), and selection of patients for adjuvant therapy or clinical trials. Could personalize care
Collapse
Affiliation(s)
- D Verver
- Department of Surgical Oncology, Erasmus MC Cancer Institute, University Medical Centre, Rotterdam, the Netherlands
| | - D van Klaveren
- Medical Statistics, Department of Biomedical Data Sciences, Leiden University Medical Centre, Leiden, the Netherlands
| | - V Franke
- Department of Surgical Oncology, Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - A C J van Akkooi
- Department of Surgical Oncology, Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - P Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Institute Cancer Centre, Warsaw, Poland
| | - U Keilholz
- Charité Comprehensive Cancer Centre, University of Medicine Berlin, Berlin, Germany
| | - A M M Eggermont
- Department of Surgical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - T Nijsten
- Department of Dermatology, Erasmus MC University Medical Centre, Rotterdam, the Netherlands
| | - D J Grünhagen
- Department of Surgical Oncology, Erasmus MC Cancer Institute, University Medical Centre, Rotterdam, the Netherlands
| | - C Verhoef
- Department of Surgical Oncology, Erasmus MC Cancer Institute, University Medical Centre, Rotterdam, the Netherlands
| |
Collapse
|
780
|
Johnson DH, Zobniw CM, Trinh VA, Ma J, Bassett RL, Abdel-Wahab N, Anderson J, Davis JE, Joseph J, Uemura M, Noman A, Abu-Sbeih H, Yee C, Amaria R, Patel S, Tawbi H, Glitza IC, Davies MA, Wong MK, Woodman S, Hwu WJ, Hwu P, Wang Y, Diab A. Infliximab associated with faster symptom resolution compared with corticosteroids alone for the management of immune-related enterocolitis. J Immunother Cancer 2018; 6:103. [PMID: 30305177 PMCID: PMC6180568 DOI: 10.1186/s40425-018-0412-0] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 09/18/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Immune-related enterocolitis (irEC) is the most common serious complication from checkpoint inhibitors (CPIs). The current front-line treatment for irEC, high-dose corticosteroids (CS), have significant side effects and prolonged therapy may reduce CPI-anti-tumor activity. Early addition of TNF-α inhibitors such as infliximab (IFX) may expedite symptom resolution and shorten CS duration. Thus, we conducted the first retrospective study, to our knowledge, evaluating symptom resolution in patients with irEC treated with and without IFX. METHODS Data were collected from the medical records of patients diagnosed with irEC. The primary endpoint was time to symptom resolution for irEC for cases managed with IFX plus CS (IFX group) versus CS alone (CS group). Duration of CS, overall survival (OS), and time to treatment failure (TTF) were secondary endpoints. RESULTS Among 75 patients with irEC, 52% received CS alone, and 48% received IFX. Despite higher grade colitis in the IFX group (grade 3/4: 86% vs. 34%; p < 0.001), median times to diarrhea resolution (3 vs. 9 days; p < 0.001) and to steroid titration (4 vs. 13 days; p < 0.001) were shorter in the IFX group than in the CS group without a negative impact on TTF or OS. Total steroid duration (median 35 vs. 51 days; p = 0.150) was numerically lower in the IFX group. CONCLUSIONS Despite higher incidence of grade 3/4 colitis, IFX added to CS for the treatment of patients with irEC was associated with a significantly shorter time to symptom resolution. The data suggest that early introduction of IFX should be considered for patients with irEC until definitive prospective clinical trials are conducted.
Collapse
Affiliation(s)
- Daniel H Johnson
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chrystia M Zobniw
- Department of Rheumatology and Rehabilitation, Faculty of Medicine, Assiut University Hospitals, Assiut, Egypt
| | - Van A Trinh
- Department of Rheumatology and Rehabilitation, Faculty of Medicine, Assiut University Hospitals, Assiut, Egypt
| | - Junsheng Ma
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Roland L Bassett
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Noha Abdel-Wahab
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Rheumatology and Rehabilitation, Faculty of Medicine, Assiut University Hospitals, Assiut, Egypt
| | - Jaime Anderson
- Department of Rheumatology and Rehabilitation, Faculty of Medicine, Assiut University Hospitals, Assiut, Egypt
| | - Jennifer E Davis
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jocelyn Joseph
- Department of Rheumatology and Rehabilitation, Faculty of Medicine, Assiut University Hospitals, Assiut, Egypt
| | - Marc Uemura
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ali Noman
- Department of Gastroenterology, Hepatology, and Nutrition, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hamzah Abu-Sbeih
- Department of Gastroenterology, Hepatology, and Nutrition, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cassian Yee
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rodabe Amaria
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sapna Patel
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hussein Tawbi
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Isabella C Glitza
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael A Davies
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael K Wong
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Scott Woodman
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wen-Jen Hwu
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Patrick Hwu
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yinghong Wang
- Department of Gastroenterology, Hepatology, and Nutrition, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Adi Diab
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
781
|
Bersanelli M, Leonetti A, Buti S. The link between calcitriol and anticancer immunotherapy: vitamin D as the possible balance between inflammation and autoimmunity in the immune-checkpoint blockade. Immunotherapy 2018; 9:1127-1131. [PMID: 29067883 DOI: 10.2217/imt-2017-0127] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Melissa Bersanelli
- Medical Oncology Unit, University Hospital of Parma, Via Gramsci 14, 43126, Parma (PR), Italy
| | - Alessandro Leonetti
- Medical Oncology Unit, University Hospital of Parma, Via Gramsci 14, 43126, Parma (PR), Italy
| | - Sebastiano Buti
- Medical Oncology Unit, University Hospital of Parma, Via Gramsci 14, 43126, Parma (PR), Italy
| |
Collapse
|
782
|
Neoadjuvant versus adjuvant ipilimumab plus nivolumab in macroscopic stage III melanoma. Nat Med 2018; 24:1655-1661. [PMID: 30297911 DOI: 10.1038/s41591-018-0198-0] [Citation(s) in RCA: 625] [Impact Index Per Article: 89.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 08/06/2018] [Indexed: 12/25/2022]
Abstract
Adjuvant ipilimumab (anti-CTLA-4) and nivolumab (anti-PD-1) both improve relapse-free survival of stage III melanoma patients1,2. In stage IV disease, the combination of ipilimumab + nivolumab is superior to ipilimumab alone and also appears to be more effective than nivolumab monotherapy3. Preclinical work suggests that neoadjuvant application of checkpoint inhibitors may be superior to adjuvant therapy4. To address this question and to test feasibility, 20 patients with palpable stage III melanoma were 1:1 randomized to receive ipilimumab 3 mg kg-1 and nivolumab 1 mg kg-1, as either four courses after surgery (adjuvant arm) or two courses before surgery and two courses postsurgery (neoadjuvant arm). Neoadjuvant therapy was feasible, with all patients undergoing surgery at the preplanned time point. However in both arms, 9/10 patients experienced one or more grade 3/4 adverse events. Pathological responses were achieved in 7/9 (78%) patients treated in the neoadjuvant arm. None of these patients have relapsed so far (median follow-up, 25.6 months). We found that neoadjuvant ipilimumab + nivolumab expand more tumor-resident T cell clones than adjuvant application. While neoadjuvant therapy appears promising, with the current regimen it induced high toxicity rates; therefore, it needs further investigation to preserve efficacy but reduce toxicity.
Collapse
|
783
|
Hieken TJ, Kane JM, Wong SL. The Role of Completion Lymph Node Dissection for Sentinel Lymph Node-Positive Melanoma. Ann Surg Oncol 2018; 26:1028-1034. [PMID: 30284132 DOI: 10.1245/s10434-018-6812-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Indexed: 12/12/2022]
Abstract
PURPOSE AND METHODS Completion lymph node dissection (CLND) for sentinel lymph node (SLN)-positive melanoma patients has been guideline-concordant standard of care since adoption of lymphatic mapping and SLN biopsy for the management of clinically node-negative melanoma patients more than 20 years ago. However, a trend for omission of CLND has been observed over the past decade, and we now have randomized, controlled clinical trial data to help guide treatment recommendations. Publication of these data prompted an American Society of Clinical Oncology-Society of Surgical Oncology 2018 clinical practice guideline update for these patients. RESULTS AND CONCLUSIONS Systematic review of current evidence supports a selective, individualized approach to CLND for SLN-positive melanoma. For low-risk, low-volume micrometastatic disease, SLN biopsy may be both diagnostic and therapeutic, and close clinical follow-up with imaging or CLND are reasonable options for appropriately selected patients. For higher-risk patients, omission of CLND requires careful consideration of risks versus benefits, relevant histopathology, and individualized patient discussion. This should address patient comorbidities and life expectancy, the predicted likelihood of additional positive nodes, availability of imaging surveillance, likelihood of adherence to imaging and clinical follow-up, consequences of regional recurrence, and the prognostic value of complete nodal staging and its impact on adjuvant therapy recommendations or clinical trial participation. Data on long-term outcomes, cost, and patient-reported quality of life measures are not yet available.
Collapse
Affiliation(s)
- Tina J Hieken
- Department of Surgery, Mayo Clinic, Rochester, MN, USA.
| | - John M Kane
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Sandra L Wong
- Department of Surgery, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| |
Collapse
|
784
|
Richtig G, Hoeller C, Wolf M, Wolf I, Rainer BM, Schulter G, Richtig M, Grübler MR, Gappmayer A, Haidn T, Kofler J, Huegel R, Lange-Asschenfeldt B, Pichler M, Pilz S, Heinemann A, Richtig E. Body mass index may predict the response to ipilimumab in metastatic melanoma: An observational multi-centre study. PLoS One 2018; 13:e0204729. [PMID: 30273398 PMCID: PMC6166940 DOI: 10.1371/journal.pone.0204729] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 09/13/2018] [Indexed: 11/18/2022] Open
Abstract
Introduction Immunotherapy is a well-established treatment option in patients with metastatic melanoma. However, biomarkers that can be used to predict a response in these patients have not yet been found, putting patients at risk of severe side effects. Methods In this retrospective analysis, we investigated the association between the body mass index and ipilimumab treatment response in patients with metastatic melanoma. Patients with metastatic melanoma who received a monotherapy of up to 4 doses of ipilimumab (3 mg/kg) every 3 weeks from 2011 to 2014 in three major hospitals in Austria were included. Patients were classified into two groups: normal group (BMI<25) and overweight group (BMI≥25). Results 40 patients had a normal BMI, and 36 had a BMI above normal. Patients with a BMI that was above normal showed significantly higher response rates (p = 0.024, χ2), and lower likelihood of brain metastases (p = 0.012, χ2). No differences were found between both groups with respect to gender (p = 0.324, χ2), T-stage (p = 0.197, χ2), or the occurrence of side effects (p = 0.646, χ2). Patients with a BMI above normal showed a trend towards longer overall survival (p = 0.056, Log-Rank), but no difference was found regarding progression-free survival (p = 0.924, Log-Rank). Conclusions The BMI correlated with the response to ipilimumab treatment in a cohort of metastatic melanoma patients.
Collapse
Affiliation(s)
- Georg Richtig
- Otto Loewi Research Center, Pharmacology Section, Medical University of Graz, Graz, Austria
- Department of Dermatology, Medical University of Graz, Graz, Austria
- * E-mail:
| | - Christoph Hoeller
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Martin Wolf
- Department of Dermatology, Medical University of Graz, Graz, Austria
- Department of Dermatology, State Hospital Wiener Neustadt, Wiener Neustadt, Austria
| | - Ingrid Wolf
- Department of Dermatology, Medical University of Graz, Graz, Austria
| | - Barbara M. Rainer
- Department of Dermatology, Medical University of Graz, Graz, Austria
| | - Günter Schulter
- Department of Psychology, Biological Psychology Unit, University of Graz, Graz, Austria
| | - Markus Richtig
- Department of Dermatology, Medical University of Graz, Graz, Austria
| | - Martin R. Grübler
- Swiss Cardiovascular Center Bern, Department of Cardiology, Bern University Hospital, Bern, Switzerland
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, Graz, Austria
| | - Anna Gappmayer
- Department of Dermatology, State Hospital Wiener Neustadt, Wiener Neustadt, Austria
| | - Thomas Haidn
- Department of Dermatology, State Hospital Wiener Neustadt, Wiener Neustadt, Austria
| | - Julian Kofler
- Department of Dermatology and Venereology, State Hospital Klagenfurt, Klagenfurt am Wörthersee, Austria
| | - Rainer Huegel
- Department of Dermatology and Venereology, State Hospital Klagenfurt, Klagenfurt am Wörthersee, Austria
| | - Bernhard Lange-Asschenfeldt
- Department of Dermatology and Venereology, State Hospital Klagenfurt, Klagenfurt am Wörthersee, Austria
- Skin Cancer Center Charité, Department of Dermatology and Allergy, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Martin Pichler
- Division of Oncology, Medical University Graz, Graz, Austria
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Stefan Pilz
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, Graz, Austria
| | - Akos Heinemann
- Otto Loewi Research Center, Pharmacology Section, Medical University of Graz, Graz, Austria
| | - Erika Richtig
- Department of Dermatology, Medical University of Graz, Graz, Austria
| |
Collapse
|
785
|
van Willigen WW, Bloemendal M, Gerritsen WR, Schreibelt G, de Vries IJM, Bol KF. Dendritic Cell Cancer Therapy: Vaccinating the Right Patient at the Right Time. Front Immunol 2018; 9:2265. [PMID: 30327656 PMCID: PMC6174277 DOI: 10.3389/fimmu.2018.02265] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/11/2018] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoint inhibitors propelled the field of oncology with clinical responses in many different tumor types. Superior overall survival over chemotherapy has been reported in various metastatic cancers. Furthermore, prolonged disease-free and overall survival have been reported in the adjuvant treatment of stage III melanoma. Unfortunately, a substantial portion of patients do not obtain a durable response. Therefore, additional strategies for the treatment of cancer are still warranted. One of the numerous options is dendritic cell vaccination, which employs the central role of dendritic cells in activating the innate and adaptive immune system. Over the years, dendritic cell vaccination was shown to be able to induce an immunologic response, to increase the number of tumor infiltrating lymphocytes and to provide overall survival benefit for at least a selection of patients in phase II studies. However, with the success of immune checkpoint inhibition in several malignancies and considering the plethora of other treatment modalities being developed, it is of utmost importance to delineate the position of dendritic cell therapy in the treatment landscape of cancer. In this review, we address some key questions regarding the integration of dendritic cell vaccination in future cancer treatment paradigms.
Collapse
Affiliation(s)
- Wouter W van Willigen
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, Netherlands.,Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
| | - Martine Bloemendal
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, Netherlands.,Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
| | - Winald R Gerritsen
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Gerty Schreibelt
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
| | - I Jolanda M de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
| | - Kalijn F Bol
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, Netherlands.,Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
| |
Collapse
|
786
|
Challenges and Opportunities of Neoadjuvant Treatment in Locally Advanced Melanoma. Am J Clin Dermatol 2018; 19:639-646. [PMID: 30039289 DOI: 10.1007/s40257-018-0371-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Locally advanced and metastatic melanoma have historically had poor survival outcomes. Long-term follow-up of both targeted therapies and immune checkpoint inhibitors has confirmed the survival benefit of these agents in stage IV melanoma, and recent studies have now demonstrated relapse-free survival benefits from these targeted and immunotherapeutic agents in the adjuvant setting. Neoadjuvant treatment of locally advanced melanoma, including in-transit disease, is now under investigation. Clinical trials have shown early promising results using either combination targeted therapy or immune checkpoint inhibitors. Neoadjuvant treatment may improve surgical morbidity, but balancing treatment efficacy and toxicity has already been challenging in the use of combination immune checkpoint inhibitors preoperatively. While improvement in relapse-free survival has been noted, additional follow-up of patients receiving neoadjuvant treatment will be necessary to report on long-term outcomes. Neoadjuvant treatment also provides additional translational research opportunities to determine predictive biomarkers for targeted therapy and immune checkpoint inhibitors. Evidence of early resistance to treatment may also lead to novel combination therapies to explore in future clinical trials. While neoadjuvant treatment in locally advanced melanoma has exciting potential, more investigation is necessary to determine efficacious regimens with manageable toxicities.
Collapse
|
787
|
Albertini MR, Yang RK, Ranheim EA, Hank JA, Zuleger CL, Weber S, Neuman H, Hartig G, Weigel T, Mahvi D, Henry MB, Quale R, McFarland T, Gan J, Carmichael L, Kim K, Loibner H, Gillies SD, Sondel PM. Pilot trial of the hu14.18-IL2 immunocytokine in patients with completely resectable recurrent stage III or stage IV melanoma. Cancer Immunol Immunother 2018; 67:1647-1658. [PMID: 30073390 PMCID: PMC6168354 DOI: 10.1007/s00262-018-2223-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 07/30/2018] [Indexed: 01/13/2023]
Abstract
Phase I testing of the hu14.18-IL2 immunocytokine (IC) in melanoma patients showed immune activation, reversible toxicities, and a maximal tolerated dose of 7.5 mg/m2/day. Preclinical data in IC-treated tumor-bearing mice with low tumor burden documented striking antitumor effects. Patients with completely resectable recurrent stage III or stage IV melanoma were scheduled to receive 3 courses of IC at 6 mg/m2/day i.v. on days 1, 2 and 3 of each 28-day course. Patients were randomized to complete surgical resection either following neoadjuvant (Group A) or prior to adjuvant (Group B) IC course 1. Primary objectives were to: (1) evaluate histological evidence of anti-tumor activity and (2) evaluate recurrence-free survival (RFS) and OS. Twenty melanoma patients were randomized to Group A (11 patients) or B (9 patients). Two Group B patients did not receive IC due to persistent disease following surgery. Six of 18 IC-treated patients remained free of recurrence, with a median RFS of 5.7 months (95% confidence interval (CI) 1.8-not reached). The 24-month RFS rate was 38.9% (95% CI 17.5-60.0%). The median follow-up of surviving patients was 50.0 months (range: 31.8-70.4). The 24-month OS rate was 65.0% (95% CI 40.3-81.5%). Toxicities were similar to those previously reported. Exploratory tumor-infiltrating lymphocyte (TIL) analyses suggest prognostic value of TILs from Group A patients. Prolonged tumor-free survival was seen in some melanoma patients at high risk for recurrence who were treated with IC.
Collapse
Affiliation(s)
- Mark R Albertini
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
- Medical Service, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
- University of Wisconsin Clinical Sciences Center, Room K6/530, 600 Highland Avenue, Madison, WI, 53792, USA.
| | - Richard K Yang
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Erik A Ranheim
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Jacquelyn A Hank
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Cindy L Zuleger
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sharon Weber
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Heather Neuman
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Greg Hartig
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Tracey Weigel
- Westchester Medical Center, New York Medical College, Valhalla, New York, USA
| | - David Mahvi
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Mary Beth Henry
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Renae Quale
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Thomas McFarland
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Jacek Gan
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Lakeesha Carmichael
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - KyungMann Kim
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | | | | | - Paul M Sondel
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
788
|
Bhutiani N, Egger ME, Stromberg AJ, Gershenwald JE, Ross MI, Philips P, Martin RCG, Scoggins CR, McMasters KM. A model for predicting low probability of nonsentinel lymph node positivity in melanoma patients with a single positive sentinel lymph node. J Surg Oncol 2018; 118:922-927. [PMID: 30259521 DOI: 10.1002/jso.25193] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 07/09/2018] [Indexed: 11/09/2022]
Abstract
BACKGROUND Identifying factors associated with nonsentinel lymph node (NSN) metastases in melanoma patients with a single positive sentinel lymph node (SLN) could aid decision making regarding adjuvant therapy. We describe a model for identifying patients with a single positive SLN at low risk for NSN metastasis. METHODS Factors associated with NSN metastasis in patients with a primary cutaneous melanoma and a single positive SLN who underwent completion lymph node dissection (CLND) were identified. These factors were used to construct a model for predicting the NSN status. The model was validated using a separate data set from another tertiary referral cancer center. RESULTS In the training data set, 111 patients had a single positive SLN. Of these, 27 had positive NSN. SLN tumor deposit diameter ≥0.75 mm (OR, 3.43; P = 0.047), age ≥40 (OR, 12.14; P = 0.024), and multifocal SLN tumor deposit location (OR, 4.16; P = 0.0096) were independently associated with NSN positivity. Patients with 0 to 1 of these risk factors had a low risk of NSN metastasis in both the training (7.5%) and validation (4.6%) data sets. CONCLUSIONS A combination of patient and SLN tumor burden characteristics can help to identify patients with a single positive SLN who are at a low risk of NSN metastasis.
Collapse
Affiliation(s)
- Neal Bhutiani
- Department of Surgery, University of Louisville, Louisville, Kentucky
| | - Michael E Egger
- Department of Surgery, University of Louisville, Louisville, Kentucky
| | | | - Jeffrey E Gershenwald
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Merrick I Ross
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Prejesh Philips
- Department of Surgery, University of Louisville, Louisville, Kentucky
| | - Robert C G Martin
- Department of Surgery, University of Louisville, Louisville, Kentucky
| | | | - Kelly M McMasters
- Department of Surgery, University of Louisville, Louisville, Kentucky
| |
Collapse
|
789
|
Kamath SD, Kumthekar PU. Immune Checkpoint Inhibitors for the Treatment of Central Nervous System (CNS) Metastatic Disease. Front Oncol 2018; 8:414. [PMID: 30319977 PMCID: PMC6171475 DOI: 10.3389/fonc.2018.00414] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 09/07/2018] [Indexed: 12/21/2022] Open
Abstract
While the CNS has long been viewed as an immune-privileged environment, a paradigm shift in neuro-immunology has elevated the role of systemic immunotherapy for the treatment of metastatic disease. Increasing knowledge regarding the presence of a CNS lymphatic system and the physical and biochemical alteration of the blood brain barrier (BBB) by the tumor microenvironment suggests immune cell trafficking in and out of the CNS is possible. Emerging clinical data suggest immune checkpoint inhibitors (ICIs) can stimulate T cells peripherally to in turn have anti-tumor effects in the CNS. For example, anti-programmed cell death-1 (PD-1) monotherapy with pembrolizumab has shown intracranial response rates of 20-30% in patients with melanoma or non-small cell lung cancer (NSCLC) brain metastases. The combination of nivolumab and ipilimumab [anti-PD-1 and anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4)] showed an intracranial response rate of 55% in patients with melanoma brain metastases. More data are needed to confirm these response rates and to determine mechanisms of efficacy and resistance. While local therapies such as stereotactic radiosurgery (SRS), whole-brain radiation therapy (WBRT), and surgery remain current mainstays, ICIS offer potential decreased neurotoxicity. This review summarizes the biological rationale for systemic immunotherapy to treat CNS metastatic disease, existing clinical data on ICIs in this setting and ongoing clinical trials exploring areas of unmet need.
Collapse
Affiliation(s)
- Suneel D. Kamath
- Northwestern Medicine, Chicago, IL, United States
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Priya U. Kumthekar
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, United States
| |
Collapse
|
790
|
Mollanoori H, Shahraki H, Rahmati Y, Teimourian S. CRISPR/Cas9 and CAR-T cell, collaboration of two revolutionary technologies in cancer immunotherapy, an instruction for successful cancer treatment. Hum Immunol 2018; 79:876-882. [PMID: 30261221 DOI: 10.1016/j.humimm.2018.09.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 09/23/2018] [Accepted: 09/23/2018] [Indexed: 12/18/2022]
Abstract
Clustered regularly interspaced short palindromic repeats/CRISPR associated nuclease9 (CRISPR/Cas9) technology, an acquired immune system in bacteria and archaea, has provided a new tool for accurately genome editing. Using only a single nuclease protein in complex with 2 short RNA as a site-specific endonuclease made it a simple and flexible genome editing tool to target nearly any genomic locus. Due to recent developments in therapeutic engineered T cell and effective responses of CD19-directed chimeric antigen receptor T cells (CART19) in patients with B-cell leukemia and lymphoma, adoptive T cell immunotherapy, particularly CAR-T cell therapy became a rapidly growing field in cancer therapy and recently Kymriah and Yescarta (CD19-directed CAR-T cells) were approved by FDA. Therefore, the combination of CRISPR/Cas9 technology as a genome engineering tool and CAR-T cell therapy (engineered T cells that express chimeric antigen receptors) may lead to further improvement in efficiency and safety of CAR-T cells. This article reviews mechanism and therapeutic application of CRISPR/Cas9 technology, accuracy of this technology, cancer immunotherapy by CAR T cells, the application of CRISPR technology for the production of universal CAR T cells, improving their antitumor efficacy, and biotech companies that invested in CRISPR technology for CAR-T cell therapy.
Collapse
Affiliation(s)
- Hasan Mollanoori
- Department of Medical Genetics, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Hojat Shahraki
- Cellular and Molecular Research Center, Zahedan University of Medical Sciences, Zahedan, Iran; Department of Laboratory Sciences, School of Allied Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Yazdan Rahmati
- Department of Medical Genetics, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Shahram Teimourian
- Department of Medical Genetics, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| |
Collapse
|
791
|
Mylvaganam GH, Chea LS, Tharp GK, Hicks S, Velu V, Iyer SS, Deleage C, Estes JD, Bosinger SE, Freeman GJ, Ahmed R, Amara RR. Combination anti-PD-1 and antiretroviral therapy provides therapeutic benefit against SIV. JCI Insight 2018; 3:122940. [PMID: 30232277 PMCID: PMC6237231 DOI: 10.1172/jci.insight.122940] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 08/17/2018] [Indexed: 01/12/2023] Open
Abstract
Therapeutic strategies that augment antiviral immunity and reduce the viral reservoir are critical to achieving durable remission of HIV. The coinhibitory receptor programmed death-1 (PD-1) regulates CD8+ T cell dysfunction during chronic HIV and SIV infections. We previously demonstrated that in vivo blockade of PD-1 during chronic SIV infection improves the function of antiviral CD8+ T cells and B cells. Here, we tested the immunological and virological effects of PD-1 blockade combined with antiretroviral therapy (ART) in rhesus macaques. Administration of anti-PD-1 antibody 10 days prior to ART initiation rapidly enhanced antiviral CD8+ T cell function and diminished IFN-stimulated genes. This resulted in faster viral suppression in plasma and better Th17 cell reconstitution in the rectal mucosa following ART initiation. PD-1 blockade during ART resulted in lower levels of cell-associated replication-competent virus. Following ART interruption, PD-1 antibody-treated animals showed markedly higher expansion of proliferating CXCR5+perforin+granzyme B+ effector CD8+ T cells and lower regulatory T cells that resulted in better control of viremia. Our results show that PD-1 blockade can be administered safely with ART to augment antiviral CD8+ T cell function and reduce the viral reservoir, leading to improved control of viral rebound after ART interruption.
Collapse
Affiliation(s)
- Geetha H. Mylvaganam
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
- Division of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Lynette S. Chea
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
- Division of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Gregory K. Tharp
- Division of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Sakeenah Hicks
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
- Division of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Vijayakumar Velu
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
- Division of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Smita S. Iyer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
- Division of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Claire Deleage
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland, USA
| | - Jacob D. Estes
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland, USA
| | - Steven E. Bosinger
- Division of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Gordon J. Freeman
- Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Rafi Ahmed
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
- Division of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Rama R. Amara
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
- Division of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
792
|
Acral Melanoma: A Patient's Experience and Physician's Commentary. Dermatol Ther (Heidelb) 2018; 8:503-507. [PMID: 30229459 PMCID: PMC6261119 DOI: 10.1007/s13555-018-0260-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Indexed: 11/01/2022] Open
Abstract
This article, co-authored by a patient diagnosed with acral melanoma, discusses the patient's experience of being diagnosed with and treated with surgery for this disease. The physician discusses the epidemiology, genetics, diagnosis, treatment, and prognosis of acral melanoma. Follow-up care plans are also discussed.
Collapse
|
793
|
Masoud SJ, Perone JA, Farrow NE, Mosca PJ, Tyler DS, Beasley GM. Sentinel Lymph Node Biopsy and Completion Lymph Node Dissection for Melanoma. Curr Treat Options Oncol 2018; 19:55. [PMID: 30232648 PMCID: PMC6684152 DOI: 10.1007/s11864-018-0575-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OPINION STATEMENT This review critically evaluates recent trials which have challenged the practice of completion lymph node dissection (CLND) for melanoma patients diagnosed with regional metastasis by positive sentinel lymph node biopsy (SLNB). Two trials in the last 2 years, DeCOG-SLT and MSLT-II, found no significant differences in melanoma-specific survival between patients, whether they received immediate CLND or observation after positive SLNB, despite decreases in nodal recurrence achieved by dissection. These trials together disfavor routine CLND in most patients after positive SLNB. However, their conclusions are limited by study populations which overall harbored a lower burden of SLN disease. Special attention needs to be given to patients who do have higher risk disease, with SLN tumor burdens exceeding 1 mm in diameter, for whom CLND may remain both prognostic and therapeutic. Current guidelines thus recommend either CLND or careful observation after positive SLNB after appropriate risk stratification of patients. While a decline in CLND is inevitable, treatment of stage III melanoma is witnessing the concurrent rise of effective adjuvant therapies. PD-1 inhibitors such as nivolumab, or combination BRAF/MEK inhibitors for V600E or K mutant melanoma, which were previously available to only trial patients with completely resected stage III disease, are now approved for use in patients with positive SLNB alone. Providers are better equipped than ever to treat clinically occult, regional metastatic disease with SLNB followed by adjuvant therapy for most patients, but should take steps to avoid undertreatment of high-risk patients who may proceed to disease relapse or progression.
Collapse
Affiliation(s)
- Sabran J Masoud
- Department of Surgery, Duke University, Durham, NC, 27710, USA
| | - Jennifer A Perone
- Department of Surgery, University Texas Medical Branch, Galveston, TX, USA
| | - Norma E Farrow
- Department of Surgery, Duke University, Durham, NC, 27710, USA
| | - Paul J Mosca
- Department of Surgery, Duke University, Durham, NC, 27710, USA
| | - Douglas S Tyler
- Department of Surgery, University Texas Medical Branch, Galveston, TX, USA
| | - Georgia M Beasley
- Department of Surgery, Duke University, Durham, NC, 27710, USA.
- Duke University Medical Center, DUMC Box 3118, Durham, NC, 27710, USA.
| |
Collapse
|
794
|
Schadendorf D, van Akkooi ACJ, Berking C, Griewank KG, Gutzmer R, Hauschild A, Stang A, Roesch A, Ugurel S. Melanoma. Lancet 2018; 392:971-984. [PMID: 30238891 DOI: 10.1016/s0140-6736(18)31559-9] [Citation(s) in RCA: 957] [Impact Index Per Article: 136.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 06/07/2018] [Accepted: 06/29/2018] [Indexed: 02/08/2023]
Abstract
Cutaneous melanoma causes 55 500 deaths annually. The incidence and mortality rates of the disease differ widely across the globe depending on access to early detection and primary care. Once melanoma has spread, this type of cancer rapidly becomes life-threatening. For more than 40 years, few treatment options were available, and clinical trials during that time were all unsuccessful. Over the past 10 years, increased biological understanding and access to innovative therapeutic substances have transformed advanced melanoma into a new oncological model for treating solid cancers. Treatments that target B-Raf proto-oncogene serine/threonine-kinase (BRAF)V600 (Val600) mutations using selected BRAF inhibitors combined with mitogen-activated protein kinase inhibitors have significantly improved response and overall survival. Furthermore, advanced cutaneous melanoma has developed into a prototype for testing checkpoint-modulating agents, which has increased hope for long-term tumour containment and a potential cure. These expectations have been sustained by clinical success with targeted agents and antibodies that block programmed cell-death protein 1 in locoregional disease, which induces prolongation of relapse-free, distant-metastasis-free, and overall survival times.
Collapse
Affiliation(s)
- Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Essen, Germany; German Cancer Consortium, Heidelberg, Germany.
| | - Alexander C J van Akkooi
- Department of Surgical Oncology, Netherlands Cancer Institute Antoni van Leeuwenhoek, Amsterdam, Netherlands
| | - Carola Berking
- Department of Dermatology and Allergy, University Hospital Munich, Munich, Germany
| | - Klaus G Griewank
- Department of Dermatology, University Hospital Essen, Essen, Germany; German Cancer Consortium, Heidelberg, Germany
| | - Ralf Gutzmer
- Department of Dermatology, Hannover Medical School, Skin Cancer Centre Hannover, Hannover, Germany
| | - Axel Hauschild
- Department of Dermatology, University Hospital, Kiel, Germany
| | - Andreas Stang
- Centre of Clinical Epidemiology, Institute of Medical Informatics, Biometry, and Epidemiology, University Hospital Essen, Essen, Germany; Department of Epidemiology, School of Public Health, Boston University, Boston, MA, USA
| | - Alexander Roesch
- Department of Dermatology, University Hospital Essen, Essen, Germany; German Cancer Consortium, Heidelberg, Germany
| | - Selma Ugurel
- Department of Dermatology, University Hospital Essen, Essen, Germany; German Cancer Consortium, Heidelberg, Germany
| |
Collapse
|
795
|
Festino L, Vanella V, Trojaniello C, Ascierto PA. Selecting immuno-oncology–based drug combinations – what should we be considering? Expert Rev Clin Pharmacol 2018; 11:971-985. [DOI: 10.1080/17512433.2018.1518713] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Lucia Festino
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | - Vito Vanella
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | - Claudia Trojaniello
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | - Paolo A. Ascierto
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| |
Collapse
|
796
|
The impact of effective systemic therapies on surgery for stage IV melanoma. Eur J Cancer 2018; 103:24-31. [PMID: 30196107 DOI: 10.1016/j.ejca.2018.08.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 08/02/2018] [Indexed: 01/11/2023]
Abstract
INTRODUCTION The outcomes of patients with metastatic melanoma have significantly improved with the introduction of effective systemic therapies (ESTs). The role of surgery in the context of ESTs for stage IV melanoma is evolving. We sought to characterise the changing patterns of surgery and oncological outcomes in patients with stage IV melanoma treated before and after the establishment of ESTs. METHODS Patients undergoing surgical resection of stage IV melanoma were identified from our institutional database from 2003 to 2015. Patients were grouped into two cohorts, those referred before EST (2003-2007) and after EST (2011-2015). Clinicopathological variables, patterns of surgery and oncological outcomes in the two groups were compared. RESULTS A total of 138 patients underwent surgery for stage IV melanoma (n = 69 in each cohort). We observed no significant difference in the ratio of operations/patients performed. However, the pattern of operations altered, with a significant decrease in in-transit excisions (0.9% vs. 19.4%, p < 0.001) and an increase in abdominal metastasectomies (21.1% vs. 4.2%, p < 0.001), in the after-EST cohort. Novel indications for surgical intervention were noted in the after-EST cohort, with a significant increase in potentially curative operations for residual oligometastatic disease (15.9% vs. 4.3%, p = 0.045). Survival after surgery was prolonged in the after-EST cohort (median 16 months vs. 6 months, p < 0.001), with the stage at initial metastasectomy (stage 4a, hazard ratio [HR] 0.45 (0.28-0.73), p = 0.001) and treatment with immune checkpoint inhibitors (HR 0.38 (0.25-0.60), p < 0.001) associated with prolonged survival. DISCUSSION Surgery remains important in the management of stage IV melanoma, with evolving indications and patterns of intervention after the introduction of ESTs. The combination of judicious surgery and EST may improve oncological outcomes.
Collapse
|
797
|
Lee JS, Adler L, Karathia H, Carmel N, Rabinovich S, Auslander N, Keshet R, Stettner N, Silberman A, Agemy L, Helbling D, Eilam R, Sun Q, Brandis A, Malitsky S, Itkin M, Weiss H, Pinto S, Kalaora S, Levy R, Barnea E, Admon A, Dimmock D, Stern-Ginossar N, Scherz A, Nagamani SCS, Unda M, Wilson DM, Elhasid R, Carracedo A, Samuels Y, Hannenhalli S, Ruppin E, Erez A. Urea Cycle Dysregulation Generates Clinically Relevant Genomic and Biochemical Signatures. Cell 2018; 174:1559-1570.e22. [PMID: 30100185 PMCID: PMC6225773 DOI: 10.1016/j.cell.2018.07.019] [Citation(s) in RCA: 215] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 05/21/2018] [Accepted: 07/12/2018] [Indexed: 01/02/2023]
Abstract
The urea cycle (UC) is the main pathway by which mammals dispose of waste nitrogen. We find that specific alterations in the expression of most UC enzymes occur in many tumors, leading to a general metabolic hallmark termed "UC dysregulation" (UCD). UCD elicits nitrogen diversion toward carbamoyl-phosphate synthetase2, aspartate transcarbamylase, and dihydrooratase (CAD) activation and enhances pyrimidine synthesis, resulting in detectable changes in nitrogen metabolites in both patient tumors and their bio-fluids. The accompanying excess of pyrimidine versus purine nucleotides results in a genomic signature consisting of transversion mutations at the DNA, RNA, and protein levels. This mutational bias is associated with increased numbers of hydrophobic tumor antigens and a better response to immune checkpoint inhibitors independent of mutational load. Taken together, our findings demonstrate that UCD is a common feature of tumors that profoundly affects carcinogenesis, mutagenesis, and immunotherapy response.
Collapse
Affiliation(s)
- Joo Sang Lee
- Cancer Data Science Lab, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Center for Bioinformatics and Computational Biology, University of Maryland Institute for Advanced Computer Studies, Department of Computer Science, University of Maryland, College Park, MD 20742, USA
| | - Lital Adler
- Department of Biological Regulation, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Hiren Karathia
- Center for Bioinformatics and Computational Biology, University of Maryland Institute for Advanced Computer Studies, Department of Computer Science, University of Maryland, College Park, MD 20742, USA
| | - Narin Carmel
- Department of Biological Regulation, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Shiran Rabinovich
- Department of Biological Regulation, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Noam Auslander
- Cancer Data Science Lab, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Center for Bioinformatics and Computational Biology, University of Maryland Institute for Advanced Computer Studies, Department of Computer Science, University of Maryland, College Park, MD 20742, USA
| | - Rom Keshet
- Department of Biological Regulation, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Noa Stettner
- Department of Biological Regulation, Weizmann Institute of Science, 7610001 Rehovot, Israel; Department of Veterinary Resources, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Alon Silberman
- Department of Biological Regulation, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Lilach Agemy
- Department of Plant and Environmental Science, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | | | - Raya Eilam
- Department of Veterinary Resources, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Qin Sun
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alexander Brandis
- Life Sciences Core Facilities, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Sergey Malitsky
- Life Sciences Core Facilities, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Maxim Itkin
- Life Sciences Core Facilities, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Hila Weiss
- Department of Biological Regulation, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Sivan Pinto
- Department of Biological Regulation, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Shelly Kalaora
- Department of Molecular Cell Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Ronen Levy
- Department of Molecular Cell Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Eilon Barnea
- Faculty of Biology, Technion - Israel Institute of Technology, 3200003 Haifa, Israel
| | - Arie Admon
- Faculty of Biology, Technion - Israel Institute of Technology, 3200003 Haifa, Israel
| | - David Dimmock
- Rady Children's Institute for Genomic Medicine, San Diego, CA 92123, USA
| | - Noam Stern-Ginossar
- Department of Molecular Genetics, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Avigdor Scherz
- Department of Veterinary Resources, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Sandesh C S Nagamani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Texas Children's Hospital, Houston, TX 77030, USA
| | - Miguel Unda
- Department of Urology, Basurto University Hospital, 48013 Bilbao, Spain; CIBERONC, Madrid, Spain
| | - David M Wilson
- Laboratory of Molecular Gerontology, National Institute on Aging, Intramural Research Program, NIH, 251 Bayview Blvd., Baltimore, MD 21224, USA
| | - Ronit Elhasid
- Sackler Faculty of Medicine, Department of Pediatric Hemato Oncology, Sourasky Medical Center, Tel Aviv University, 6997801 Tel Aviv, Israel
| | - Arkaitz Carracedo
- CIBERONC, Madrid, Spain; CIC bioGUNE, Bizkaia Technology Park, 801 Building, 48160 Derio, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain; Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - Yardena Samuels
- Department of Molecular Cell Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Sridhar Hannenhalli
- Center for Bioinformatics and Computational Biology, University of Maryland Institute for Advanced Computer Studies, Department of Computer Science, University of Maryland, College Park, MD 20742, USA
| | - Eytan Ruppin
- Cancer Data Science Lab, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Center for Bioinformatics and Computational Biology, University of Maryland Institute for Advanced Computer Studies, Department of Computer Science, University of Maryland, College Park, MD 20742, USA; Schools of Medicine and Computer Science, Tel Aviv University, 6997801 Tel Aviv, Israel.
| | - Ayelet Erez
- Department of Biological Regulation, Weizmann Institute of Science, 7610001 Rehovot, Israel.
| |
Collapse
|
798
|
Smith H, Wilkinson M, Smith M, Strauss D, Hayes A. The effect of age on outcomes after isolated limb perfusion for advanced extremity malignancies. Eur J Cancer 2018; 100:46-54. [DOI: 10.1016/j.ejca.2018.04.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 04/05/2018] [Accepted: 04/11/2018] [Indexed: 12/14/2022]
|
799
|
Hansen ED, Wang X, Case AA, Puzanov I, Smith T. Immune Checkpoint Inhibitor Toxicity Review for the Palliative Care Clinician. J Pain Symptom Manage 2018; 56:460-472. [PMID: 29792979 DOI: 10.1016/j.jpainsymman.2018.05.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 05/15/2018] [Accepted: 05/15/2018] [Indexed: 12/22/2022]
Abstract
Immune checkpoint inhibitors (ICIs) have opened an exciting chapter in the treatment of patients with advanced cancer. For the palliative care clinician, however, ICIs present several new challenges, including new ways to define treatment success, as well as treatment-related toxicities that differ in nature and timing from traditional chemotherapy. In this article, we review the mechanism of action of ICIs, as well as selected published data supporting the efficacy of ICIs in patients with advanced cancer. In addition, we summarize existing data of ICI toxicity prevalence, patterns of severity, and timing of onset. Finally, we briefly review key principles from published guidelines on the management of ICI toxicities.
Collapse
Affiliation(s)
- Eric D Hansen
- Department of Supportive Care, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA.
| | - Xiao Wang
- Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Amy A Case
- Department of Supportive Care, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Igor Puzanov
- Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Tom Smith
- Palliative Medicine Program, Johns Hopkins Hospital, Baltimore, Maryland, USA
| |
Collapse
|
800
|
Combinatory therapy adopting nanoparticle-based cancer vaccination with immune checkpoint blockade for treatment of post-surgical tumor recurrences. J Control Release 2018; 285:56-66. [DOI: 10.1016/j.jconrel.2018.07.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 07/05/2018] [Accepted: 07/05/2018] [Indexed: 12/20/2022]
|