1
|
Koban MU, Hartmann M, Amexis G, Franco P, Huggins L, Shah I, Karachaliou N. Targeted Therapies, Novel Antibodies, and Immunotherapies in Advanced Non-Small Cell Lung Cancer: Clinical Evidence and Drug Approval Patterns. Clin Cancer Res 2024; 30:4822-4833. [PMID: 39177967 PMCID: PMC11528205 DOI: 10.1158/1078-0432.ccr-24-0741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/23/2024] [Accepted: 07/11/2024] [Indexed: 08/24/2024]
Abstract
Since 2011, the US FDA has approved 30 new drugs for use in advanced non-small cell lung cancer (NSCLC), mainly comprising tyrosine kinase inhibitors and immune checkpoint inhibitors. NSCLC with oncogene driver alterations is amenable to treatment with targeted drugs, usually small-molecule inhibitors. In these cases, the demonstration of high overall response rates, coupled with a lasting duration of response, has allowed for accelerated approval in the United States, based on single-cohort or multicohort trials. Confirmatory clinical evidence was subsequently provided through postmarketing trials. In NSCLC without such driver alterations, regulatory agencies in both the United States and the European Union set clinical evidence expectations that foster the conduct of studies primarily focused on determining survival or event-free survival, based on randomized controlled trial designs. This review analyzes the approval patterns of novel therapeutics for NSCLC with a focus on small-molecule inhibitors that target driver alterations, as well as biologics. The latter include mAbs inhibiting immune checkpoints like PD-(L)1 or cell surface receptors and antibody-drug conjugates, highly potent biologics linked to a cytotoxic compound. The differentiation of NSCLC into oncogene- and non-oncogene-addicted subtypes determines drug development strategies, the extent of the clinical development program, access to orphan drug development incentives, and regulatory approval strategies.
Collapse
Affiliation(s)
- Marén U. Koban
- Global Regulatory and Scientific Policy, The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | | | - Georgios Amexis
- Global Regulatory Affairs Oncology, The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | - Pedro Franco
- Merck Serono Limited UK, an Affiliate of Merck KGaA, Feltham, United Kingdom
| | - Laura Huggins
- Global Regulatory Affairs Oncology, The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | | | - Niki Karachaliou
- Global Clinical Development, The Healthcare Business of Merck KGaA, Darmstadt, Germany
| |
Collapse
|
2
|
Serrano P, Wah Yuen H, Akdemir J, Hartmann M, Reinholz T, Peltier S, Ligensa T, Seiller C, Paraiso Le Bourhis A. Real-world data in drug development strategies for orphan drugs: tafasitamab in B cell lymphoma, a case study for approval based on a single-arm combination trial. Drug Discov Today 2022; 27:1706-1715. [PMID: 35218926 DOI: 10.1016/j.drudis.2022.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/31/2022] [Accepted: 02/19/2022] [Indexed: 12/01/2022]
Abstract
Tafasitamab (TAF) plus lenalidomide (LEN) is a novel treatment option for patients with relapsed/refractory diffuse large B cell lymphoma (rrDLBCL) who are not eligible for autologous stem cell transplantation. The initial US/EU approvals for TAF represent precedents because this is the first time that approval of a novel combination therapy was granted based on a pivotal single-arm trial (SAT). Matching real world-data (RWD) helped to disentangle the contribution of individual agents. In this review, we present the TAF development strategy, the prospective incorporation of RWD within the clinical development plan, the corresponding regulatory hurdles of this strategy, and the prior regulatory actions for other cancer drugs that previously incorporated RWD and propensity score matching in EU and US regulatory submissions. We also outline how RWD could further advance and impact orphan drug development.
Collapse
Affiliation(s)
| | | | | | - Markus Hartmann
- European Consulting & Contracting in Oncology, Trier, Germany
| | | | | | | | | | | |
Collapse
|
3
|
Sebastian M, Gröschel A, Gütz S, Schulz H, Müller-Huesmann H, Liersch R, von der Heyde E, Wiegand J, Ukena D, Bargon J, Schütte W, Riera-Knorrenschild J, Fischer JR, Griesinger F, Allan V, Waldenberger D, Schumann C. Prospective, Noninterventional Study of Nivolumab in Real-world Patients With Locally Advanced or Metastatic Non-small Cell Lung Cancer After Prior Chemotherapy (ENLARGE-Lung). J Immunother 2022; 45:89-99. [PMID: 34908007 DOI: 10.1097/cji.0000000000000397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 05/18/2021] [Indexed: 11/26/2022]
Abstract
Nivolumab was the first immune checkpoint inhibitor approved for use in advanced non-small cell lung cancer (NSCLC). This noninterventional, prospective cohort study investigates real-world effectiveness of nivolumab in pretreated NSCLC patients in Germany (Enlarge-Lung/CA209-580). Patients with squamous (SQ) or nonsquamous (NSQ) NSCLC previously treated for locally advanced or metastatic (stage IIIB/IV) disease received nivolumab according to the current Summary of Product Characteristics. Overall survival (OS) was the primary endpoint. Of 907 patients enrolled, 660 patients who were followed for at least 12 months across 79 study centers in Germany, were analyzed. Median OS was 11.2 months [95% confidence interval (CI), 9.1-12.9]; outcomes for the 418 patients with NSQ histology [13.1 mo (95% CI, 10.6-15.6)] were more favorable than outcomes for the 242 patients with SQ histology [8.9 mo (95% CI, 6.4-11.3)]. Patients' age, presence of distant or brain metastases, or line of therapy did not affect outcomes; however, patients with poor performance status (ECOG-PS ≥2, n=80) had shorter median OS [4.7 mo (95% CI, 3.1-5.4)]. This study represents one of the largest real-world cohorts providing outcomes of nivolumab in pretreated NSCLC. The results match well with the published evidence from pivotal clinical trials and demonstrate clinical effectiveness of nivolumab in advanced NSCLC.
Collapse
Affiliation(s)
| | - Andreas Gröschel
- Department of Internal Medicine II-Pneumology & Respiratory Medicine, Clemenshospital
| | - Sylvia Gütz
- Department of Pneumology and Cardiology, Ev. Diakonissenkrankenhaus gGmbH, Leipzig
| | - Holger Schulz
- Practice for Hematology and Oncology, PIOH Frechen, Frechen
| | | | | | | | - Jörg Wiegand
- Practice for Hematology and Oncology, St. Josef Hospital, Moers
| | - Dieter Ukena
- Department for Pneumology and Respiratory Medicine, Klinikum Bremen-Ost, Bremen
| | - Joachim Bargon
- Department for Pneumology, Frankfurter Rotkreuz-Kliniken, Frankfurt, Germany
| | - Wolfgang Schütte
- Department of Internal Medicine, Krankenhaus Martha Maria Halle Dölau gGmbH, Halle
| | | | | | - Frank Griesinger
- Department of Hematology and Oncology, Pius-Hospital, University Department Internal Medicine-Oncology, University Medicine Oldenburg, Germany
| | - Victoria Allan
- Center for Observational Research & Data Science, Bristol-Myers Squibb, Uxbridge, UK
| | | | - Christian Schumann
- Clinic for Pneumology, Thoracic Oncology, Sleep and Respiratory Medicine, Klinikverbund Allgäu gGmbH, Kempten and Immenstadt, Germany
| |
Collapse
|
4
|
de Jong G, Bartels L, Kedde M, Verdegaal EME, Gillissen MA, Levie SE, Cercel MG, van Hal-van Veen SE, Fatmawati C, van de Berg D, Yasuda E, Claassen YB, Bakker AQ, van der Burg SH, Schotte R, Villaudy J, Spits H, Hazenberg MD, van Helden PM, Wagner K. Melanoma cells can be eliminated by sialylated CD43 × CD3 bispecific T cell engager formats in vitro and in vivo. Cancer Immunol Immunother 2020; 70:1569-1581. [PMID: 33225419 DOI: 10.1007/s00262-020-02780-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 10/22/2020] [Indexed: 01/13/2023]
Abstract
Targeted cancer therapy with monoclonal antibodies has proven successful for different cancer types but is limited by the availability of suitable antibody targets. CD43s, a unique sialylated form of CD43 expressed by hematologic malignancies, is a recently identified target and antibodies interacting with CD43s may have therapeutic potential against acute myeloid leukemia (AML) and myelodysplastic syndrome. CD43s is recognized by the human antibody AT1413, that was derived from a high-risk AML patient who successfully cleared leukemia after allogeneic stem cell transplantation. Here we observed that AT1413 binds also to certain non-hematopoietic tumor cells, particularly melanoma and breast cancer. AT1413 immune precipitated CD43s from melanoma cells confirming that it recognizes the same target on melanoma as on AML. AT1413 induced antibody-dependent cellular cytotoxicity against short-term cultured patient-derived melanoma samples. However, AT1413 was unable to affect the growth of melanoma cells in vivo. To increase the efficacy of AT1413 as a therapeutic antibody, we generated two different formats of bispecific T-cell engaging antibodies (TCEs): one binding bivalently (bTCE) and the other monovalently (knob-in-hole; KiH) to both CD43s and CD3ε. In vitro, these TCEs redirected T-cell cytotoxicity against melanoma cells with differences in potencies. To investigate their effects in vivo, we grafted mice that harbor a human immune system with the melanoma cell line A375. Treatment with both AT1413 bTCE and AT1413 KiH significantly reduced tumor outgrowth in these mice. These data indicate a broad therapeutic potential of AT1413 that includes AML and CD43s-expressing solid tumors that originate from CD43-negative tissues.
Collapse
Affiliation(s)
- G de Jong
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands.,Department of Hematology, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands.,Cancer Center Amsterdam (CCA), Amsterdam, The Netherlands.,Amsterdam Infection and Immunity Institute (AI&II), Amsterdam, The Netherlands
| | - L Bartels
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands.,Amsterdam Infection and Immunity Institute (AI&II), Amsterdam, The Netherlands
| | - M Kedde
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| | - E M E Verdegaal
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - M A Gillissen
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands.,Department of Hematology, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands.,Cancer Center Amsterdam (CCA), Amsterdam, The Netherlands.,Amsterdam Infection and Immunity Institute (AI&II), Amsterdam, The Netherlands
| | - S E Levie
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| | - M G Cercel
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| | | | - C Fatmawati
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| | - D van de Berg
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| | - E Yasuda
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| | - Y B Claassen
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| | - A Q Bakker
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| | - S H van der Burg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - R Schotte
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| | - J Villaudy
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| | - H Spits
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands.,Amsterdam Infection and Immunity Institute (AI&II), Amsterdam, The Netherlands.,Department of Experimental Immunology, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands
| | - M D Hazenberg
- Department of Hematology, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands.,Cancer Center Amsterdam (CCA), Amsterdam, The Netherlands.,Amsterdam Infection and Immunity Institute (AI&II), Amsterdam, The Netherlands.,Department of Hematopoiesis, Sanquin Research, Amsterdam, The Netherlands
| | - P M van Helden
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands.
| | - K Wagner
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| |
Collapse
|
5
|
Regev A, Avigan MI, Kiazand A, Vierling JM, Lewis JH, Omokaro SO, Di Bisceglie AM, Fontana RJ, Bonkovsky HL, Freston JW, Uetrecht JP, Miller ED, Pehlivanov ND, Haque SA, Harrison MJ, Kullak-Ublick GA, Li H, Patel NN, Patwardhan M, Price KD, Watkins PB, Chalasani NP. Best practices for detection, assessment and management of suspected immune-mediated liver injury caused by immune checkpoint inhibitors during drug development. J Autoimmun 2020; 114:102514. [DOI: 10.1016/j.jaut.2020.102514] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 07/07/2020] [Indexed: 02/07/2023]
|
6
|
Dimou A, Grewe P, Sidney J, Sette A, Norman PJ, Doebele RC. HLA Class I Binding of Mutant EGFR Peptides in NSCLC Is Associated With Improved Survival. J Thorac Oncol 2020; 16:104-112. [PMID: 32927123 DOI: 10.1016/j.jtho.2020.08.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 07/11/2020] [Accepted: 08/30/2020] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Cancer-associated mutations have the potential to generate neoantigens and elicit CD8-positive T-cell-dependent adaptive immune responses. There are currently no reports of CD8-positive T-cells with specificity for neoepitopes generated by EGFR mutations, which are driver oncogenes in a subset of patients with lung cancer. METHODS We used NETMHCpan 4.0 to identify putative protective human leukocyte antigen (HLA) class I allotypes that are predicted in silico to bind and present mutant EGFR-generated peptides on the basis of predefined criteria. We associated the presence or absence of these alleles with clinical outcomes in patients from The Cancer Genome Atlas with lung adenocarcinoma. RESULTS We identified 12 HLA class I alleles that fulfilled the predefined criteria for being protective for EGFR p.L858R and six for EGFR p.E746_A750del, the two most common EGFR mutations in lung cancer. We validated the in silico predictions for peptide-HLA allele binding in vitro. A third (12 of 36) of patients with mostly early stage lung adenocarcinoma in The Cancer Genome Atlas with either EGFR p.L858R or EGFR p.E746_A750del had at least one protective allele in their host genomes. More importantly, patients with protective alleles exhibited better disease-free (hazard ratio: 0.20, 95% confidence interval: 0.05-0.78) and overall survival (hazard ratio: 0.13, 95% confidence interval: 0.02-0.64), and this effect was independent of the EGFR mutation type, stage, age, and sex. CONCLUSIONS Our data revealed that clinical outcomes were improved in patients with EGFR mutation-positive lung adenocarcinoma who harbored protective HLA class I alleles. Thus, immunity with specificity for mutant EGFR is possible in a subset of patients with early stage lung cancer and portends a better prognosis.
Collapse
Affiliation(s)
- Anastasios Dimou
- Division of Medical Oncology, School of Medicine, University of Colorado, Aurora, Colorado; Department of Medical Oncology, Mayo Clinic, Rochester, Minnesota.
| | - Paul Grewe
- Division of Medical Oncology, School of Medicine, University of Colorado, Aurora, Colorado
| | - John Sidney
- La Jolla Institute for Immunology, La Jolla, California
| | - Alessandro Sette
- La Jolla Institute for Immunology, La Jolla, California; Department of Medicine, University of California in San Diego, La Jolla, California
| | - Paul J Norman
- Division of Biomedical Informatics and Personalized Medicine, School of Medicine, University of Colorado, Aurora, Colorado; Department of Microbiology and Immunology, School of Medicine, University of Colorado, Aurora, Colorado
| | - Robert C Doebele
- Division of Medical Oncology, School of Medicine, University of Colorado, Aurora, Colorado
| |
Collapse
|
7
|
Bullement A, Willis A, Amin A, Schlichting M, Hatswell AJ, Bharmal M. Evaluation of survival extrapolation in immuno-oncology using multiple pre-planned data cuts: learnings to aid in model selection. BMC Med Res Methodol 2020; 20:103. [PMID: 32375680 PMCID: PMC7204248 DOI: 10.1186/s12874-020-00997-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 04/28/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Due to limited duration of follow up in clinical trials of cancer treatments, estimates of lifetime survival benefits are typically derived using statistical extrapolation methods. To justify the method used, a range of approaches have been proposed including statistical goodness-of-fit tests and comparing estimates against a previous data cut (i.e. interim data collected). In this study, we extend these approaches by presenting a range of extrapolations fitted to four pre-planned data cuts from the JAVELIN Merkel 200 (JM200) trial. By comparing different estimates of survival and goodness-of-fit as JM200 data mature, we undertook an iterative process of fitting and re-fitting survival models to retrospectively identify early indications of likely long-term survival. METHODS Standard and spline-based parametric models were fitted to overall survival data from each JM200 data cut. Goodness-of-fit was determined using an assessment of the estimated hazard function, information theory-based methods and objective comparisons of estimation accuracy. Best-fitting extrapolations were compared to establish which one provided the most accurate estimation, and how statistical goodness-of-fit differed. RESULTS Spline-based models provided the closest fit to the final JM200 data cut, though all extrapolation methods based on the earliest data cut underestimated the 'true' long-term survival (difference in restricted mean survival time [RMST] at 36 months: - 1.1 to - 0.5 months). Goodness-of-fit scores illustrated that an increasingly flexible model was favored as data matured. Given an early data cut, a more flexible model better aligned with clinical expectations could be reasonably justified using a range of metrics, including RMST and goodness-of-fit scores (which were typically within a 2-point range of the statistically 'best-fitting' model). CONCLUSIONS Survival estimates from the spline-based models are more aligned with clinical expectation and provided a better fit to the JM200 data, despite not exhibiting the definitively 'best' statistical goodness-of-fit. Longer-term data are required to further validate extrapolations, though this study illustrates the importance of clinical plausibility when selecting the most appropriate model. In addition, hazard-based plots and goodness-of-fit tests from multiple data cuts present useful approaches to identify when a more flexible model may be advantageous. TRIAL REGISTRATION JAVELIN Merkel 200 was registered with ClinicalTrials.gov as NCT02155647 on June 4, 2014.
Collapse
Affiliation(s)
| | | | | | | | - Anthony James Hatswell
- Delta Hat, Nottingham, UK
- Department of Statistical Science, University College London, London, UK
| | - Murtuza Bharmal
- Oncology Brands & Life Cycle Management, Global Evidence & Value Development, EMD Serono, Inc, One Technology Place, Rockland, MA, 02370, USA.
| |
Collapse
|
8
|
Dubey D, David WS, Reynolds KL, Chute DF, Clement NF, Cohen JV, Lawrence DP, Mooradian MJ, Sullivan RJ, Guidon AC. Severe Neurological Toxicity of Immune Checkpoint Inhibitors: Growing Spectrum. Ann Neurol 2020; 87:659-669. [PMID: 32086972 DOI: 10.1002/ana.25708] [Citation(s) in RCA: 128] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 12/11/2022]
Abstract
Expanding use of immune-checkpoint inhibitors (ICIs) underscores the importance of accurate diagnosis and timely management of neurological immune-related adverse events (irAE-N). We evaluate the real-world frequency, phenotypes, co-occurring immune-related adverse events (irAEs), and long-term outcomes of severe, grade III to V irAE-N at a tertiary care center over 6 years. We analyze how our experience supports published literature and professional society guidelines. We also discuss these data with regard to common clinical scenarios, such as combination therapy, ICI rechallenge and risk of relapse of irAE-N, and corticosteroid taper, which are not specifically addressed by current guidelines and/or have limited data. Recommendations for management and future irAE-N reporting are outlined. ANN NEUROL 2020;87:659-669.
Collapse
Affiliation(s)
- Divyanshu Dubey
- Department of Neurology, Massachusetts General Hospital, Boston, MA.,Department of Neurology, Brigham and Women's Hospital, Boston, MA.,Department of Neurology, and Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - William S David
- Department of Neurology, Massachusetts General Hospital, Boston, MA
| | - Kerry L Reynolds
- Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Donald F Chute
- Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Nathan F Clement
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - Justine V Cohen
- Department of Medicine, Massachusetts General Hospital, Boston, MA
| | | | | | - Ryan J Sullivan
- Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Amanda C Guidon
- Department of Neurology, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
9
|
Sidders B, Zhang P, Goodwin K, O'Connor G, Russell DL, Borodovsky A, Armenia J, McEwen R, Linghu B, Bendell JC, Bauer TM, Patel MR, Falchook GS, Merchant M, Pouliot G, Barrett JC, Dry JR, Woessner R, Sachsenmeier K. Adenosine Signaling Is Prognostic for Cancer Outcome and Has Predictive Utility for Immunotherapeutic Response. Clin Cancer Res 2020; 26:2176-2187. [PMID: 31953314 DOI: 10.1158/1078-0432.ccr-19-2183] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 11/15/2019] [Accepted: 01/14/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE There are several agents in early clinical trials targeting components of the adenosine pathway including A2AR and CD73. The identification of cancers with a significant adenosine drive is critical to understand the potential for these molecules. However, it is challenging to measure tumor adenosine levels at scale, thus novel, clinically tractable biomarkers are needed. EXPERIMENTAL DESIGN We generated a gene expression signature for the adenosine signaling using regulatory networks derived from the literature and validated this in patients. We applied the signature to large cohorts of disease from The Cancer Genome Atlas (TCGA) and cohorts of immune checkpoint inhibitor-treated patients. RESULTS The signature captures baseline adenosine levels in vivo (r 2 = 0.92, P = 0.018), is reduced after small-molecule inhibition of A2AR in mice (r 2 = -0.62, P = 0.001) and humans (reduction in 5 of 7 patients, 70%), and is abrogated after A2AR knockout. Analysis of TCGA confirms a negative association between adenosine and overall survival (OS, HR = 0.6, P < 2.2e-16) as well as progression-free survival (PFS, HR = 0.77, P = 0.0000006). Further, adenosine signaling is associated with reduced OS (HR = 0.47, P < 2.2e-16) and PFS (HR = 0.65, P = 0.0000002) in CD8+ T-cell-infiltrated tumors. Mutation of TGFβ superfamily members is associated with enhanced adenosine signaling and worse OS (HR = 0.43, P < 2.2e-16). Finally, adenosine signaling is associated with reduced efficacy of anti-PD1 therapy in published cohorts (HR = 0.29, P = 0.00012). CONCLUSIONS These data support the adenosine pathway as a mediator of a successful antitumor immune response, demonstrate the prognostic potential of the signature for immunotherapy, and inform patient selection strategies for adenosine pathway modulators currently in development.
Collapse
Affiliation(s)
- Ben Sidders
- Translational Medicine, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, United Kingdom.
| | - Pei Zhang
- Translational Medicine, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Kelly Goodwin
- Discovery, Research and Early Development, Oncology R&D, AstraZeneca, Boston, Massachusetts
| | - Greg O'Connor
- Translational Medicine, Research and Early Development, Oncology R&D, AstraZeneca, Boston, Massachusetts
| | - Deanna L Russell
- Translational Medicine, Research and Early Development, Oncology R&D, AstraZeneca, Boston, Massachusetts
| | - Alexandra Borodovsky
- Discovery, Research and Early Development, Oncology R&D, AstraZeneca, Boston, Massachusetts
| | - Joshua Armenia
- Translational Medicine, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Robert McEwen
- Translational Medicine, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Bolan Linghu
- Translational Medicine, Research and Early Development, Oncology R&D, AstraZeneca, Boston, Massachusetts
| | - Johanna C Bendell
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, Tennessee
| | - Todd M Bauer
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, Tennessee
| | - Manish R Patel
- Sarah Cannon Research Institute/Florida Cancer Specialists, Sarasota, Florida
| | | | - Melinda Merchant
- Early Clinical Development, Research and Early Development, Oncology R&D, AstraZeneca, Boston, Massachusetts
| | - Gayle Pouliot
- Early Clinical Development, Research and Early Development, Oncology R&D, AstraZeneca, Boston, Massachusetts
| | - J Carl Barrett
- Translational Medicine, Research and Early Development, Oncology R&D, AstraZeneca, Boston, Massachusetts
| | - Jonathan R Dry
- Translational Medicine, Research and Early Development, Oncology R&D, AstraZeneca, Boston, Massachusetts
| | - Rich Woessner
- Discovery, Research and Early Development, Oncology R&D, AstraZeneca, Boston, Massachusetts
| | - Kris Sachsenmeier
- Translational Medicine, Research and Early Development, Oncology R&D, AstraZeneca, Boston, Massachusetts
| |
Collapse
|