Clinical activity of single-dose systemic oncolytic VSV virotherapy in patients with relapsed refractory T-cell lymphoma

Joselle Cook, Kah-Whye Peng, Thomas E Witzig, Stephen M Broski, Jose C Villasboas, Jonas Paludo, Mrinal Patnaik, Vincent Rajkumar, Angela Dispenzieri, Nelson Leung, Francis Buadi, Nora Bennani, Stephen M Ansell, Lianwen Zhang, Nandakumar Packiriswamy, Baskar Balakrishnan, Bethany Brunton, Marissa Giers, Brenda Ginos, Amylou C Dueck, Susan Geyer, Morie A Gertz, Rahma Warsame, Ronald S Go, Suzanne R Hayman, David Dingli, Shaji Kumar, Leif Bergsagel, Javier L Munoz, Wilson Gonsalves, Taxiarchis Kourelis, Eli Muchtar, Prashant Kapoor, Robert A Kyle, Yi Lin, Mustaqeem Siddiqui, Amie Fonder, Miriam Hobbs, Lisa Hwa, Shruthi Naik, Stephen J Russell, Martha Q Lacy, Joselle Cook, Kah-Whye Peng, Thomas E Witzig, Stephen M Broski, Jose C Villasboas, Jonas Paludo, Mrinal Patnaik, Vincent Rajkumar, Angela Dispenzieri, Nelson Leung, Francis Buadi, Nora Bennani, Stephen M Ansell, Lianwen Zhang, Nandakumar Packiriswamy, Baskar Balakrishnan, Bethany Brunton, Marissa Giers, Brenda Ginos, Amylou C Dueck, Susan Geyer, Morie A Gertz, Rahma Warsame, Ronald S Go, Suzanne R Hayman, David Dingli, Shaji Kumar, Leif Bergsagel, Javier L Munoz, Wilson Gonsalves, Taxiarchis Kourelis, Eli Muchtar, Prashant Kapoor, Robert A Kyle, Yi Lin, Mustaqeem Siddiqui, Amie Fonder, Miriam Hobbs, Lisa Hwa, Shruthi Naik, Stephen J Russell, Martha Q Lacy

Abstract

Clinical success with intravenous (IV) oncolytic virotherapy (OV) has to-date been anecdotal. We conducted a phase 1 clinical trial of systemic OV and investigated the mechanisms of action in responding patients. A single IV dose of vesicular stomatitis virus (VSV) interferon-β (IFN-β) with sodium iodide symporter (NIS) was administered to patients with relapsed/refractory hematologic malignancies to determine safety and efficacy across 4 dose levels (DLs). Correlative studies were undertaken to evaluate viremia, virus shedding, virus replication, and immune responses. Fifteen patients received VSV-IFNβ-NIS. Three patients were treated at DL1 through DL3 (0.05, 0.17, and 0.5 × 1011 TCID50), and 6 were treated at DL4 (1.7 × 1011 TCID50) with no dose-limiting toxicities. Three of 7 patients with T-cell lymphoma (TCL) had responses: a 3-month partial response (PR) at DL2, a 6-month PR, and a complete response (CR) ongoing at 20 months at DL4. Viremia peaked at the end of infusion, g was detected. Plasma IFN-β, a biomarker of VSV-IFNβ-NIS replication, peaked between 4 hours and 48 hours after infusion. The patient with CR had robust viral replication with increased plasma cell-free DNA, high peak IFN-β of 18 213 pg/mL, a strong anti-VSV neutralizing antibody response, and increased numbers of tumor reactive T-cells. VSV-IFNβ-NIS as a single agent was effective in patients with TCL, resulting in durable disease remissions in heavily pretreated patients. Correlative analyses suggest that responses may be due to a combination of direct oncolytic tumor destruction and immune-mediated tumor control. This trial is registered at www.clinicaltrials.gov as #NCT03017820.

© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.

Figures

Graphical abstract
Graphical abstract
Figure 1.
Figure 1.
Clinical outcomes of treatment with 1 IV infusion of VSV-IFNβ-NIS. (A) Summary of patients treated in the study. (B) Maximum grade AEs by DL for each patient. (C) Levels of selected cytokines/chemokines before and after VSV infusion. (D) Swimmer plot (n = 15), in which each bar represents an individual patient as designated. Day 28 responses are indicated. Bars with solid arrows show patients with an ongoing response. ULOQ, upper limit of quantitation.
Figure 2.
Figure 2.
A single dose of VSV-IFNβ-NIS monotherapy has clinical activity in patients with TCL. Photographs (A) and PET/CT scans (B) showing resolution of FDG-avid lesions of patient 4A.3 with TCL with cutaneous relapse of nodal PTCL who achieved a CR after receiving 1 dose of VSV-IFNβ-NIS at DL4. (C) FDG PET images of patient 4A.4 with nodal PTCL showing remarkable improvement in FDG-avid cervical, mediastinal, axillary, and abdominal lymph node disease (arrows) with decreased splenic size and FDG uptake (arrowheads) at 1 month. (D) Coronal fused FDG PET/CT images demonstrate improvement in splenic disease (arrowheads), with continued response in the spleen at the 6-month evaluation.
Figure 3.
Figure 3.
Positive NIS imaging of virus-infected tumor corresponded with reduction in FDG avidity. (A) In patient 3A.1 with multiple myeloma, axial fused 99mTc-pertechnetate SPECT/CT images demonstrate increasing uptake within a lytic lesion in the anterior right ilium (arrows) on comparison at baseline before VSV, day 1 (24 hours), and day 5 images (arrows). (B) This same lesion demonstrated decreased FDG activity when comparing pre- and posttherapy PET/CT images (arrows). (C) In contrast, a left acetabular lesion in the same patient demonstrated no increased uptake on SPECT/CT at baseline, day 1, or day 5 scans, suggesting minimal VSV infection (arrows). The 6-month follow-up CT (D) and fused PET/CT images (E) showed progression of the acetabular lesion, with increased size, bone destruction, and FDG uptake (arrows).
Figure 4.
Figure 4.
Pharmacokinetics and pharmacodynamics of VSV-IFNβ-NIS after IV infusion. (A) Profile of viremia (VSV-N RNA in blood) in each patient measured at baseline, end of infusion, 30 minutes, 60 minutes, 4 hours, 24 hours, and 3, 8, 15, and 28 days after VSV. (B) 24-hour viremia levels of each patient at all DLs. Corresponding plasma IFN-β profile (C) and peak IFN-β levels (D) per DL. (E) Cell-free DNA in plasma of patients. (F) IFN-γ ELISPOT assay for T cells reactive against select tumor antigens, VSV-N, or cytomegalovirus, Epstein-Barr virus, and Flu virus positive control. (G) Anti-VSV neutralizing antibody titers (average of patients per DL). Mean ± standard error of the mean shown where applicable.

References

    1. Russell L, Peng KW, Russell SJ, Diaz RM. Oncolytic viruses: priming time for cancer immunotherapy. BioDrugs. 2019;33(5):485-501.
    1. Raja J, Ludwig JM, Gettinger SN, Schalper KA, Kim HS. Oncolytic virus immunotherapy: future prospects for oncology. J Immunother Cancer. 2018;6(1):140.
    1. Russell SJ, Peng KW, Bell JC. Oncolytic virotherapy. Nat Biotechnol. 2012;30(7):658-670.
    1. Lawler SE, Speranza MC, Cho CF, Chiocca EA. Oncolytic viruses in cancer treatment: a review. JAMA Oncol. 2017;3(6):841-849.
    1. Andtbacka RHI, Collichio F, Harrington KJ, et al. . Final analyses of OPTiM: a randomized phase III trial of talimogene laherparepvec versus granulocyte-macrophage colony-stimulating factor in unresectable stage III-IV melanoma. J Immunother Cancer. 2019;7(1):145.
    1. Greig SL. Talimogene Laherparepvec: first global approval. Drugs. 2016;76(1):147-154.
    1. Russell SJ, Federspiel MJ, Peng KW, et al. . Remission of disseminated cancer after systemic oncolytic virotherapy. Mayo Clin Proc. 2014;89(7):926-933.
    1. Pecora AL, Rizvi N, Cohen GI, et al. . Phase I trial of intravenous administration of PV701, an oncolytic virus, in patients with advanced solid cancers. J Clin Oncol. 2002;20(9):2251-2266.
    1. Maroun J, Muñoz-Alía M, Ammayappan A, Schulze A, Peng KW, Russell S. Designing and building oncolytic viruses. Future Virol. 2017;12(4): 193-213.
    1. Ferguson MS, Lemoine NR, Wang Y. Systemic delivery of oncolytic viruses: hopes and hurdles. Adv Virol. 2012;2012:805629.
    1. Miller A, Nace R, Ayala-Breton C, et al. . Perfusion pressure is a critical determinant of the intratumoral extravasation of oncolytic viruses. Mol Ther. 2016;24(2):306-317.
    1. Kueberuwa G, Cawood R, Seymour LW. Blood compatibility of enveloped viruses. Curr Opin Mol Ther. 2010;12(4):412-420.
    1. Letchworth GJ, Rodriguez LL, Del Cbarrera J. Vesicular stomatitis. Vet J. 1999;157(3):239-260.
    1. Lichty BD, Power AT, Stojdl DF, Bell JC. Vesicular stomatitis virus: re-inventing the bullet. Trends Mol Med. 2004;10(5):210-216.
    1. Felt SA, Grdzelishvili VZ. Recent advances in vesicular stomatitis virus-based oncolytic virotherapy: a 5-year update. J Gen Virol. 2017;98(12):2895-2911.
    1. Mitchell MJ, Billingsley MM, Haley RM, Wechsler ME, Peppas NA, Langer R. Engineering precision nanoparticles for drug delivery. Nat Rev Drug Discov. 2021;20(2):101-124.
    1. Naik S, Nace R, Federspiel MJ, Barber GN, Peng KW, Russell SJ. Curative one-shot systemic virotherapy in murine myeloma. Leukemia. 2012; 26(8):1870-1878.
    1. Medrano RFV, Hunger A, Mendonça SA, Barbuto JAM, Strauss BE. Immunomodulatory and antitumor effects of type I interferons and their application in cancer therapy. Oncotarget. 2017;8(41):71249-71284.
    1. Miller A, Russell SJ. The use of the NIS reporter gene for optimizing oncolytic virotherapy. Expert Opin Biol Ther. 2016;16(1):15-32.
    1. Zhang L, Steele MB, Jenks N, et al. . Safety studies in tumor and non-tumor-bearing mice in support of clinical trials using oncolytic VSV-IFNβ-NIS. Hum Gene Ther Clin Dev. 2016;27(3):111-122.
    1. Naik S, Galyon GD, Jenks NJ, et al. . Comparative oncology evaluation of intravenous recombinant oncolytic vesicular stomatitis virus therapy in spontaneous canine cancer. Mol Cancer Ther. 2018;17(1):316-326.
    1. LeBlanc AK, Naik S, Galyon GD, et al. . Safety studies on intravenous administration of oncolytic recombinant vesicular stomatitis virus in purpose-bred beagle dogs. Hum Gene Ther Clin Dev. 2013;24(4):174-181.
    1. Velazquez-Salinas L, Naik S, Pauszek SJ, Peng KW, Russell SJ, Rodriguez LL. Oncolytic recombinant vesicular stomatitis virus (VSV) is nonpathogenic and nontransmissible in pigs, a natural host of VSV. Hum Gene Ther Clin Dev. 2017;28(2):108-115.
    1. Lee DW, Gardner R, Porter DL, et al. . Current concepts in the diagnosis and management of cytokine release syndrome [published correction appears in Blood. 2015;126(8):1048]. Blood. 2014;124(2):188-195.
    1. Le Tourneau C, Lee JJ, Siu LL. Dose escalation methods in phase I cancer clinical trials. J Natl Cancer Inst. 2009;101(10):708-720.
    1. Rajkumar SV, Dimopoulos MA, Palumbo A, et al. . International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma. Lancet Oncol. 2014;15(12):e538-e548.
    1. Van Heertum RL, Scarimbolo R, Wolodzko JG, et al. . Lugano 2014 criteria for assessing FDG-PET/CT in lymphoma: an operational approach for clinical trials. Drug Des Devel Ther. 2017;11:1719-1728.
    1. Döhner H, Estey E, Grimwade D, et al. . Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2017;129(4):424-447.
    1. Lee DW, Santomasso BD, Locke FL, et al. . ASTCT consensus grading for cytokine release syndrome and neurologic toxicity associated with immune effector cells. Biol Blood Marrow Transplant. 2019;25(4):625-638.
    1. Bailey K, Kirk A, Naik S, et al. . Mathematical model for radial expansion and conflation of intratumoral infectious centers predicts curative oncolytic virotherapy parameters. PLoS One. 2013;8(9):e73759.
    1. Breitbach CJ, De Silva NS, Falls TJ, et al. . Targeting tumor vasculature with an oncolytic virus. Mol Ther. 2011;19(5):886-894.
    1. Liu YP, Suksanpaisan L, Steele MB, Russell SJ, Peng KW. Induction of antiviral genes by the tumor microenvironment confers resistance to virotherapy. Sci Rep. 2013;3(1):2375.
    1. Carey BL, Ahmed M, Puckett S, Lyles DS. Early steps of the virus replication cycle are inhibited in prostate cancer cells resistant to oncolytic vesicular stomatitis virus. J Virol. 2008;82(24):12104-12115.
    1. Holbrook MC, Goad DW, Grdzelishvili VZ. Expanding the spectrum of pancreatic cancers responsive to vesicular stomatitis virus-based oncolytic virotherapy: challenges and solutions. Cancers (Basel). 2021;13(5):1171.
    1. Baltzis D, Qu LK, Papadopoulou S, et al. . Resistance to vesicular stomatitis virus infection requires a functional cross talk between the eukaryotic translation initiation factor 2alpha kinases PERK and PKR. J Virol. 2004;78(23):12747-12761.
    1. Dummer R, Döbbeling U, Geertsen R, Willers J, Burg G, Pavlovic J. Interferon resistance of cutaneous T-cell lymphoma-derived clonal T-helper 2 cells allows selective viral replication. Blood. 2001;97(2):523-527.
    1. Tesfay MZ, Kirk AC, Hadac EM, et al. . PEGylation of vesicular stomatitis virus extends virus persistence in blood circulation of passively immunized mice. J Virol. 2013;87(7):3752-3759.
    1. Roex G, Timmers M, Wouters K, et al. . Safety and clinical efficacy of BCMA CAR-T-cell therapy in multiple myeloma. J Hematol Oncol. 2020; 13(1):164.
    1. Ollila TA, Sahin I, Olszewski AJ. Mogamulizumab: a new tool for management of cutaneous T-cell lymphoma. OncoTargets Ther. 2019;12:1085-1094.
    1. Zinzani PL, Bonthapally V, Huebner D, Lutes R, Chi A, Pileri S. Panoptic clinical review of the current and future treatment of relapsed/refractory T-cell lymphomas: peripheral T-cell lymphomas. Crit Rev Oncol Hematol. 2016;99:214-227.
    1. Jiang H, DeGrado TR. [18F]Tetrafluoroborate ([18F]TFB) and its analogs for PET imaging of the sodium/iodide symporter. Theranostics. 2018; 8(14):3918-3931.
    1. Rahmim A, Zaidi H. PET versus SPECT: strengths, limitations and challenges. Nucl Med Commun. 2008;29(3):193-207.
    1. Jiang H, Schmit NR, Koenen AR, et al. . Safety, pharmacokinetics, metabolism and radiation dosimetry of 18F-tetrafluoroborate (18F-TFB) in healthy human subjects. EJNMMI Res. 2017;7(1):90.
    1. Diocou S, Volpe A, Jauregui-Osoro M, et al. . [18F]tetrafluoroborate-PET/CT enables sensitive tumor and metastasis in vivo imaging in a sodium iodide symporter-expressing tumor model. Sci Rep. 2017;7(1):946.
    1. Zhang L, Steele MB, Jenks N, et al. . Robust oncolytic virotherapy induces tumor lysis syndrome and associated toxicities in the MPC-11 plasmacytoma model. Mol Ther. 2016;24(12):2109-2117.
    1. Sivanand A, Surmanowicz P, Alhusayen R, et al. . Immunotherapy for cutaneous T-cell lymphoma: current landscape and future developments. J Cutan Med Surg. 2019;23(5):537-544.
    1. Mehta-Shah N, Ratner L, Horwitz SM. Adult T-cell leukemia/lymphoma. J Oncol Pract. 2017;13(8):487-492.

Source: PubMed

3
Abonner