251
|
Blons H, Garinet S, Laurent-Puig P, Oudart JB. Molecular markers and prediction of response to immunotherapy in non-small cell lung cancer, an update. J Thorac Dis 2019; 11:S25-S36. [PMID: 30775025 DOI: 10.21037/jtd.2018.12.48] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Immunotherapy represents one of the most promising therapeutic approaches in lung cancer, however 50% of lung cancer patients will not respond to this treatment, while others will have transitory or durable responses. Because side effects may be life threatening and treatment costs remain very high, the identification of predictive markers is mandatory and actually extensively studied. Factors that determine response to immune checkpoint inhibitors (ICI) are numerous including tumor microenvironment, immune tumor infiltrates, expression of immune checkpoint proteins (PD-1/PD-L1), gene expression signatures and molecular tumor profiles. Based on high impact factor publications and recent literature this review focuses on the potential predictive value of tumor molecular alterations and tumor mutation burden as predictive markers of response or resistance to ICI. We also discuss the role of circulating tumor DNA (ctDNA) to monitor ICI responses and propose an algorithm that integrates molecular markers upcoming recommendations for first line treatment.
Collapse
Affiliation(s)
- Hélène Blons
- INSERM UMR-S1147, Paris Sorbonne Cite University, Paris, France.,Department of Biochemistry, Unit of Pharmacogenetics and Molecular Oncology, Georges Pompidou European Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Simon Garinet
- INSERM UMR-S1147, Paris Sorbonne Cite University, Paris, France.,Department of Biochemistry, Unit of Pharmacogenetics and Molecular Oncology, Georges Pompidou European Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Pierre Laurent-Puig
- INSERM UMR-S1147, Paris Sorbonne Cite University, Paris, France.,Department of Biochemistry, Unit of Pharmacogenetics and Molecular Oncology, Georges Pompidou European Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jean-Baptiste Oudart
- Department of Biochemistry, Unit of Pharmacogenetics and Molecular Oncology, Georges Pompidou European Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|
252
|
Passiglia F, Cappuzzo F, Alabiso O, Bettini AC, Bidoli P, Chiari R, Defferrari C, Delmonte A, Finocchiaro G, Francini G, Gelsomino F, Giannarelli D, Giordano M, Illiano A, Livi L, Martelli O, Natoli C, Puppo G, Ricevuto E, Roca E, Turci D, Galetta D. Efficacy of nivolumab in pre-treated non-small-cell lung cancer patients harbouring KRAS mutations. Br J Cancer 2019; 120:57-62. [PMID: 30377342 PMCID: PMC6325128 DOI: 10.1038/s41416-018-0234-3] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 07/12/2018] [Accepted: 07/26/2018] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The present study investigated the efficacy and safety of nivolumab in pre-treated patients with advanced NSCLC harbouring KRAS mutations. METHODS Clinical data and KRAS mutational status were analysed in patients treated with nivolumab within the Italian Expanded Access Program. Objective response rate, progression-free survival and overall survival were evaluated. Patients were monitored for adverse events using the National Cancer Institute Common Terminology Criteria for Adverse Events. RESULTS Among 530 patients evaluated for KRAS mutations, 206 (39%) were positive while 324 (61%) were KRAS wild-type mutations. KRAS status did not influence nivolumab efficacy in terms of ORR (20% vs 17%, P = 0.39) and DCR (47% vs 41%, P = 0.23). The median PFS and OS were 4 vs 3 months (P = 0.5) and 11.2 vs 10 months (P = 0.8) in the KRAS-positive vs the KRAS-negative group. The 3-months PFS rate was significantly higher in the KRAS-positive group as compared to the KRAS-negative group (53% vs 42%, P = 0.01). The percentage of any grade and grade 3-4 AEs were 45% vs 33% (P = 0.003) and 11% vs 6% (P = 0.03) in KRAS-positive and KRAS-negative groups, respectively. CONCLUSIONS Nivolumab is an effective and safe treatment option for patients with previously treated, advanced non-squamous NSCLC regardless of KRAS mutations.
Collapse
Affiliation(s)
- Francesco Passiglia
- Department of Surgical, Oncological and Stomatological Disciplines, University of Palermo, Palermo, Italy
| | - Federico Cappuzzo
- Department of Oncology and Hematology, AUSL Romagna, Ravenna, Italy.
| | - Oscar Alabiso
- University Hospital "Maggiore della Carità", Novara, Italy
| | | | | | - Rita Chiari
- Department of Medical Oncology, Santa Maria della Misericordia Hospital, University of Perugia, Perugia, Italy
| | | | - Angelo Delmonte
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumouri (IRST), Meldola, Italy
| | | | | | | | - Diana Giannarelli
- Biostatistics Unit, Scientific Direction, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | | | | | | | | - Clara Natoli
- Department of Medical, Oral and Biotechnological Sciencesl, CeSI-MeT University G. D'Annunzio, Chieti-Pescara, Italy
| | | | - Enrico Ricevuto
- Assistenza Oncologica Territoriale, Ospedale San Salvatore, Rete Oncologica ASL1 Abruzzo, Università di L'Aquila, Aquila, Italy
| | - Elisa Roca
- ASST Spedali Civili di Brescia, Brescia, Italy
| | | | | |
Collapse
|
253
|
Xiong J, Zhao WL. Advances in multiple omics of natural-killer/T cell lymphoma. J Hematol Oncol 2018; 11:134. [PMID: 30514323 PMCID: PMC6280527 DOI: 10.1186/s13045-018-0678-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 11/20/2018] [Indexed: 12/23/2022] Open
Abstract
Natural-killer/T cell lymphoma (NKTCL) represents the most common subtype of extranodal lymphoma with aggressive clinical behavior. Prevalent in Asians and South Americans, the pathogenesis of NKTCL remains to be fully elucidated. Using system biology techniques including genomics, transcriptomics, epigenomics, and metabolomics, novel biomarkers and therapeutic targets have been revealed in NKTCL. Whole-exome sequencing studies identify recurrent somatic gene mutations, involving RNA helicases, tumor suppressors, JAK-STAT pathway molecules, and epigenetic modifiers. Another genome-wide association study reports that single nucleotide polymorphisms mapping to the class II MHC region on chromosome 6 contribute to lymphomagenesis. Alterations of oncogenic signaling pathways janus kinase-signal transducer and activator of transcription (JAK-STAT), nuclear factor-κB (NF-κB), mitogen-activated protein kinase (MAPK), WNT, and NOTCH, as well as epigenetic dysregulation of microRNA and long non-coding RNAs, are also frequently observed in NKTCL. As for metabolomic profiling, abnormal amino acids metabolism plays an important role on disease progression of NKTCL. Of note, through targeting multiple omics aberrations, clinical outcome of NKTCL patients has been significantly improved by asparaginase-based regimens, immune checkpoints inhibitors, and histone deacetylation inhibitors. Future investigations will be emphasized on molecular classification of NKTCL using integrated analysis of system biology, so as to optimize targeted therapeutic strategies of NKTCL in the era of precision medicine.
Collapse
Affiliation(s)
- Jie Xiong
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, 200025, China
| | - Wei-Li Zhao
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, 200025, China. .,Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai, China.
| |
Collapse
|
254
|
Xiong D, Wang Y, Singavi AK, Mackinnon AC, George B, You M. Immunogenomic Landscape Contributes to Hyperprogressive Disease after Anti-PD-1 Immunotherapy for Cancer. iScience 2018; 9:258-277. [PMID: 30439581 PMCID: PMC6234258 DOI: 10.1016/j.isci.2018.10.021] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/14/2018] [Accepted: 10/19/2018] [Indexed: 12/11/2022] Open
Abstract
Although PD-1-blocking immunotherapies demonstrate significant therapeutic promise, a subset of the patients could develop hyperprogressive disease (HPD) with accelerated tumor growth after anti-PD1 immunotherapy. To elucidate the underlying mechanisms, we compared the mutational and transcriptional landscapes between the pre- and post-therapy tumors of two patients developing HPD after anti-PD-1 immunotherapy. In post-therapy HPD tumors, somatic mutations were found in known cancer genes, including tumor suppressor genes such as TSC2 and VHL, along with transcriptional upregulation of oncogenic pathways, including IGF-1, ERK/MAPK, PI3K/AKT, and TGF-β. We found that post-therapy HPD tumors were less immunogenic than pre-therapy tumors, concurrent with an increased presence of ILC3 cells, a subset of innate lymphoid cells. We also developed a gene expression signature predictive of HPD. In summary, we identified the genomics and immune features associated with HPD, which may help identify patients at risk of adverse clinical outcome after anti-PD-1 immunotherapy.
Collapse
Affiliation(s)
- Donghai Xiong
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Yian Wang
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Arun K Singavi
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Alexander C Mackinnon
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ben George
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ming You
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
255
|
Tuccitto A, Shahaj E, Vergani E, Ferro S, Huber V, Rodolfo M, Castelli C, Rivoltini L, Vallacchi V. Immunosuppressive circuits in tumor microenvironment and their influence on cancer treatment efficacy. Virchows Arch 2018; 474:407-420. [PMID: 30374798 DOI: 10.1007/s00428-018-2477-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 10/10/2018] [Accepted: 10/14/2018] [Indexed: 01/05/2023]
Abstract
It has been for long conceived that hallmarks of cancer were intrinsic genetic features driving tumor development, proliferation, and progression, and that targeting such cell-autonomous pathways could be sufficient to achieve therapeutic cancer control. Clinical ex vivo data demonstrated that treatment efficacy often relied on the contribution of host immune responses, hence introducing the concept of tumor microenvironment (TME), namely the existence, along with tumor cells, of non-tumor components that could significantly influence tumor growth and survival. Among the complex network of TME-driving forces, immunity plays a key role and the balance between antitumor and protumor immune responses is a major driver in contrasting or promoting cancer spreading. TME is usually a very immunosuppressed milieu because of a vast array of local alterations contrasting antitumor adaptive immunity, where metabolic changes contribute to cancer dissemination by impairing T cell infiltration and favoring the accrual and activation of regulatory cells. Subcellular structures known as extracellular vesicles then help spreading immunosuppression at systemic levels by distributing genetic and protein tumor repertoire in distant tissues. A major improvement in the knowledge of TME is now pointing the attention back to tumor cells; indeed, recent findings are showing how oncogenic pathways and specific mutations in tumor cells can actually dictate the nature and the function of immune infiltrate. As our information on the reciprocal interactions regulating TME increases, finding a strategy to interfere with TME crosstalk becomes more complex and challenging. Nevertheless, TME interactions represent a promising field for the discovery of novel biomarkers and therapeutic targets for improving treatment efficacy in cancer.
Collapse
Affiliation(s)
- Alessandra Tuccitto
- Unit of Immunotherapy of Human Tumors, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via G. Venezian 1, 20133, Milan, Italy
| | - Eriomina Shahaj
- Unit of Immunotherapy of Human Tumors, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via G. Venezian 1, 20133, Milan, Italy.
| | - Elisabetta Vergani
- Unit of Immunotherapy of Human Tumors, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via G. Venezian 1, 20133, Milan, Italy
| | - Simona Ferro
- Unit of Immunotherapy of Human Tumors, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via G. Venezian 1, 20133, Milan, Italy
| | - Veronica Huber
- Unit of Immunotherapy of Human Tumors, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via G. Venezian 1, 20133, Milan, Italy
| | - Monica Rodolfo
- Unit of Immunotherapy of Human Tumors, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via G. Venezian 1, 20133, Milan, Italy
| | - Chiara Castelli
- Unit of Immunotherapy of Human Tumors, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via G. Venezian 1, 20133, Milan, Italy
| | - Licia Rivoltini
- Unit of Immunotherapy of Human Tumors, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via G. Venezian 1, 20133, Milan, Italy
| | - Viviana Vallacchi
- Unit of Immunotherapy of Human Tumors, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via G. Venezian 1, 20133, Milan, Italy
| |
Collapse
|
256
|
Lupo A, Alifano M, Wislez M, Boulle G, Velut Y, Biton J, Cremer I, Goldwasser F, Leroy K, Damotte D. [Biomarkers predictive of PD1/PD-L1 immunotherapy in non-small cell lung cancer]. REVUE DE PNEUMOLOGIE CLINIQUE 2018; 74:339-350. [PMID: 30337201 DOI: 10.1016/j.pneumo.2018.09.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Immune checkpoint inhibitors (ICI), targeting the PD1/PD-L1 axis has shown their efficacy in lung cancer but only in a restricted population of patients, thus it is mandatory to identify biomarkers predicting the clinical benefit. In this article we will describe and analyzed biomarkers already published, from protein, to RNA and at last DNA markers, discussing each markers feasibility and interest. In the future, combined analysis of several markers will probably be proposed, particularly with the increasing complexity of therapy schema with molecules association.
Collapse
Affiliation(s)
- A Lupo
- UMRS 1138, Institut national de la santé et de la recherche médicale (Inserm), centre de recherche des cordeliers équipe « Cancer, Immune Control and Escape », Paris, France; Université Paris Descartes, 75006 Paris, France; Université Pierre et Marie-Curie Paris, 75005 Paris, France; Service de pathologie, hôpital Cochin, Assistance publique des hôpitaux de Paris (AP-HP), 75014 Paris, France
| | - M Alifano
- Université Paris Descartes, 75006 Paris, France; Service de chirurgie thoracique, hôpital Cochin, Assistance publique des hôpitaux de Paris (AP-HP), 75014 Paris, France
| | - M Wislez
- UMRS 1138, Institut national de la santé et de la recherche médicale (Inserm), centre de recherche des cordeliers équipe « Cancer, Immune Control and Escape », Paris, France; Université Paris Descartes, 75006 Paris, France; Université Pierre et Marie-Curie Paris, 75005 Paris, France; Service de pathologie, hôpital Cochin, Assistance publique des hôpitaux de Paris (AP-HP), 75014 Paris, France; Service de pneumologie, hôpital Tenon, Assistance publique des hôpitaux de Paris (AP-HP), 75014 Paris, France
| | - G Boulle
- UMRS 1138, Institut national de la santé et de la recherche médicale (Inserm), centre de recherche des cordeliers équipe « Cancer, Immune Control and Escape », Paris, France; Université Paris Descartes, 75006 Paris, France; Université Pierre et Marie-Curie Paris, 75005 Paris, France
| | - Y Velut
- UMRS 1138, Institut national de la santé et de la recherche médicale (Inserm), centre de recherche des cordeliers équipe « Cancer, Immune Control and Escape », Paris, France; Université Paris Descartes, 75006 Paris, France; Université Pierre et Marie-Curie Paris, 75005 Paris, France
| | - J Biton
- Laboratoire immunologie et immunopathologie-Li2P, Inserm UMR 1125, physiopathologie, cibles et thérapies de la polyarthrite rhumatoïde, Sorbonne Paris cité université Paris 13, 93000 Bobigny, France
| | - I Cremer
- UMRS 1138, Institut national de la santé et de la recherche médicale (Inserm), centre de recherche des cordeliers équipe « Cancer, Immune Control and Escape », Paris, France; Université Paris Descartes, 75006 Paris, France; Université Pierre et Marie-Curie Paris, 75005 Paris, France
| | - F Goldwasser
- Université Paris Descartes, 75006 Paris, France; Service d'oncologie médicale, hôpital Cochin, Assistance publique des hôpitaux de Paris (AP-HP), 75014 Paris, France
| | - K Leroy
- Université Paris Descartes, 75006 Paris, France; Service de génétique et biologie moléculaires, hôpital Cochin, Assistance publique des hôpitaux de Paris (AP-HP), 75014 Paris, France
| | - D Damotte
- UMRS 1138, Institut national de la santé et de la recherche médicale (Inserm), centre de recherche des cordeliers équipe « Cancer, Immune Control and Escape », Paris, France; Université Paris Descartes, 75006 Paris, France; Université Pierre et Marie-Curie Paris, 75005 Paris, France; Service de pathologie, hôpital Cochin, Assistance publique des hôpitaux de Paris (AP-HP), 75014 Paris, France.
| |
Collapse
|
257
|
Barta JA, McMahon SB. Lung-Enriched Mutations in the p53 Tumor Suppressor: A Paradigm for Tissue-Specific Gain of Oncogenic Function. Mol Cancer Res 2018; 17:3-9. [PMID: 30224539 DOI: 10.1158/1541-7786.mcr-18-0357] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 07/25/2018] [Accepted: 08/22/2018] [Indexed: 01/01/2023]
Abstract
Lung cancer, the leading cause of cancer-related mortality in the United States, occurs primarily due to prolonged exposure to an array of carcinogenic compounds in cigarette smoke. These carcinogens create bulky DNA adducts, inducing alterations including missense mutations in the tumor suppressor gene TP53 TP53 is the most commonly mutated gene in many human cancers, and a specific set of these variants are enriched in lung cancer (at amino acid residues V157, R158, and A159). This perspective postulates that lung-enriched mutations can be explained, in part, by biological selection for oncogenic gain-of-function (GOF) mutant p53 alleles at V157, R158, and A159. This hypothesis explaining tissue-specific TP53 mutations is further supported by mouse model studies of the canonical TP53 hotspots showing that tumor spectra and GOF activities are altered with mutation type. Therefore, although smoking-related lung cancer unequivocally arises due to the mutagenic environment induced by tobacco carcinogens, this perspective provides a rationale for the preferential selection of lung-enriched V157, R158, and A159 mutant p53.
Collapse
Affiliation(s)
- Julie A Barta
- Division of Pulmonary and Critical Care Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Steven B McMahon
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania.
| |
Collapse
|
258
|
Rochigneux P, Garon EB. Are lung adenocarcinoma mutations shaping the immune microenvironment? Transl Cancer Res 2018; 7:S740-S742. [PMID: 30906701 DOI: 10.21037/tcr.2018.07.17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Philippe Rochigneux
- Medical Oncology Department, Paoli-Calmettes Institute, 13009 Marseille, France.,Immunity & Cancer team, Inserm, U1068, Centre de Recherche en Cancérologie de Marseille, Paoli-Calmettes Institute, Aix-Marseille University, France.,David Geffen School of Medicine at the University of California, Los Angeles 90095, USA
| | - Edward B Garon
- David Geffen School of Medicine at the University of California, Los Angeles 90095, USA
| |
Collapse
|