Lymphocytic infiltration in the cutaneous lymphoma microenvironment after injection of TG1042

Nathalie Accart, Mirjana Urosevic-Maiwald, Reinhard Dummer, Vincent Bataille, Nadine Kehrer, Cristina Niculescu, Jean-Marc Limacher, Marie-Pierre Chenard, Jean-Yves Bonnefoy, Ronald Rooke, Nathalie Accart, Mirjana Urosevic-Maiwald, Reinhard Dummer, Vincent Bataille, Nadine Kehrer, Cristina Niculescu, Jean-Marc Limacher, Marie-Pierre Chenard, Jean-Yves Bonnefoy, Ronald Rooke

Abstract

Background: Primary cutaneous lymphomas (CLs), characterized by an accumulation of clonal T or B lymphocytes preferentially localized in the skin, have been successfully treated with interferons (IFNs) which counterbalance the Th2-immunosuppressive state associated with this pathology. In a phase I/II clinical trial, we correlated the local immune infiltrate and the anti-tumor effects of repeated intralesional administrations of an adenovirus vector expressing human interferon-gamma (IFN-g) termed TG1042, in patients with advanced primary cutaneous T-cell lymphomas (CTCL) or multilesional cutaneous B-cell lymphomas (CBCL).

Methods: For each patient, variation in time of specific lymphocyte populations, defined by immunohistochemical stainings, was assessed in biopsies of injected lesions. For each patient, the change in local immune response was associated with the patient's objective response at the end of the study.

Results: Immunohistochemical analyses of biopsies indicate that infiltration of CD8+ T lymphocytes and of TIA-1+ cytotoxic T-cells in lesions injected with TG1042 correlates with clinical benefit.

Conclusions: These data suggest for the first time that a CD8+ cytotoxic infiltrate, induced by local expression of IFN-g correlates with a clinical response.

Trial registration: The phase I step (TG1042.01) does not have a registration number. The phase II step (TG1042.06) registration number was NCT00394693.

Figures

Figure 1
Figure 1
Immune infiltrates in a CTCL complete responder before and after TG1042 treatment. Patient #24 at baseline (left panels) and at last visit, 4 weeks after last TG1042 injection (right panels). Percentages on pictures correspond to the ratio of DAB-positive cells on total hematoxylin-stained nuclei in the whole infiltrating population, as determined by ImageJ software macro-commands. The proportion of CD3 and CD4 expressing cells decreased at the last timepoint; the number of CD8 and CD79 α expressing cells increased between baseline and last visit.
Figure 2
Figure 2
Immune infiltrates in a CTCL progressive disease before and after TG1042 injections. Patient #27 at baseline (left panels) and at C1D16 (right panels). Percentages on pictures correspond to the ratio of DAB-positive cells on total hematoxylin-stained nuclei in the whole infiltrating population, as determined by ImageJ software macro-commands. The frequency of CD3 expressing cells increased between baseline and C1D16; CD4, CD8 and TIA-1 were stable markers. (Insert) TIA-1 staining is associated with intracellular granules.
Figure 3
Figure 3
Immune infiltrates in a CBCL complete responder before and after TG1042 injections. Patient #33 at baseline (left panels) and at C1D16 (right panels). Percentages on pictures correspond to the ratio of DAB-positive cells on total hematoxylin-stained nuclei in the whole infiltrating population, as determined by ImageJ software macro-commands. CD79α, CD20 and TIA-1 are stable markers; the frequency of CD8 expressing cells increased between baseline and C1D16.
Figure 4
Figure 4
Immune infiltrates in a CBCL partial responder before and after TG1042 injections. Patient #35 at baseline (left panels) and at C1D16 (right panels). Percentages on pictures correspond to the ratio of DAB-positive cells on total hematoxylin-stained nuclei in the whole infiltrating population, as determined by ImageJ software macro-commands. The frequency of CD79α, CD20, CD8 or TIA-1 expressing cells decreased between baseline and C1D16.

References

    1. Willemze R, Jaffe ES, Burg G, Cerroni L, Berti E, Swerdlow SH, Ralfkiaer E, Chimenti S, Diaz-Perez JL, Duncan LM. et al.WHO-EORTC classification for cutaneous lymphomas. Blood. 2005;11:3768–3785. doi: 10.1182/blood-2004-09-3502.
    1. Dummer R, Willers J, Kamarashev J, Urosevic M, Dobbeling U, Burg G. Pathogenesis of cutaneous lymphomas. Semin Cutan Med Surg. 2000;11:78–86. doi: 10.1016/S1085-5629(00)80003-6.
    1. Nagatani T, Kim S, Baba N, Miyamoto H, Minato K, Shimoyama M, Nakajima H. Phenotypic heterogeneity of lymphoma of the skin. J Dermatol. 1989;11:443–452.
    1. Olsen E, Vonderheid E, Pimpinelli N, Willemze R, Kim Y, Knobler R, Zackheim H, Duvic M, Estrach T, Lamberg S. et al.Revisions to the staging and classification of mycosis fungoides and Sezary syndrome: a proposal of the International Society for Cutaneous Lymphomas (ISCL) and the cutaneous lymphoma task force of the European Organization of Research and Treatment of Cancer (EORTC) Blood. 2007;11:1713–1722. doi: 10.1182/blood-2007-03-055749.
    1. Gilliam AC, Wood GS. Primary cutaneous lymphomas other than mycosis fungoides. Semin Oncol. 1999;11:290–306.
    1. Parks JD, Synovec MS, Masih AS, Braddock SW, Nakamine H, Sanger WG, Harrington DS, Weisenburger DD. Immunophenotypic and genotypic characterization of lymphomatoid papulosis. J Am Acad Dermatol. 1992;11:968–975. doi: 10.1016/0190-9622(92)70143-4.
    1. Beljaards RC, Meijer CJ, Van der Putte SC, Hollema H, Geerts ML, Bezemer PD, Willemze R. Primary cutaneous T-cell lymphoma: clinicopathological features and prognostic parameters of 35 cases other than mycosis fungoides and CD30-positive large cell lymphoma. J Pathol. 1994;11:53–60. doi: 10.1002/path.1711720110.
    1. Santucci M, Pimpinelli N, Arganini L. Primary cutaneous B-cell lymphoma: a unique type of low-grade lymphoma. Clinicopathologic and immunologic study of 83 cases. Cancer. 1991;11:2311–2326. doi: 10.1002/1097-0142(19910501)67:9<2311::AID-CNCR2820670918>;2-0.
    1. Yang B, Tubbs RR, Finn W, Carlson A, Pettay J, Hsi ED. Clinicopathologic reassessment of primary cutaneous B-cell lymphomas with immunophenotypic and molecular genetic characterization. Am J Surg Pathol. 2000;11:694–702. doi: 10.1097/00000478-200005000-00008.
    1. Dummer R, Goldinger SM, Cozzio A, French LE, Karpova MB. Cutaneous lymphomas: molecular pathways leading to new drugs. J Invest Dermatol. 2012;11:517–525. doi: 10.1038/jid.2011.370.
    1. Rook AH, Junkins-Hopkins JM, McGinnis KS, Wysocka M, Richardson SK, Budgin JB, Everitts S, Vittorio CC. Cytokines and other biologic agents as immunotherapeutics for cutaneous T-cell lymphoma. Adv Dermatol. 2002;11:29–43.
    1. Bunn PA Jr, Foon KA, Ihde DC, Longo DL, Eddy J, Winkler CF, Veach SR, Zeffren J, Sherwin S, Oldham R. Recombinant leukocyte A interferon: an active agent in advanced cutaneous T-cell lymphomas. Ann Intern Med. 1984;11:484–487. doi: 10.7326/0003-4819-101-4-484.
    1. Kaplan EH, Rosen ST, Norris DB, Roenigk HH Jr, Saks SR, Bunn PA Jr. Phase II study of recombinant human interferon gamma for treatment of cutaneous T-cell lymphoma. J Natl Cancer Inst. 1990;11:208–212. doi: 10.1093/jnci/82.3.208.
    1. Rook AH, Wood GS, Yoo EK, Elenitsas R, Kao DM, Sherman ML, Witmer WK, Rockwell KA, Shane RB, Lessin SR, Vonderheid EC. Interleukin-12 therapy of cutaneous T-cell lymphoma induces lesion regression and cytotoxic T-cell responses. Blood. 1999;11:902–908.
    1. Cope A, Le Friec G, Cardone J, Kemper C. The Th1 life cycle: molecular control of IFN-gamma to IL-10 switching. Trends Immunol. 2011;11:278–286. doi: 10.1016/j.it.2011.03.010.
    1. Dummer R, Hassel JC, Fellenberg F, Eichmuller S, Maier T, Slos P, Acres B, Bleuzen P, Bataille V, Squiban P. et al.Adenovirus-mediated intralesional interferon-gamma gene transfer induces tumor regressions in cutaneous lymphomas. Blood. 2004;11:1631–1638. doi: 10.1182/blood-2004-01-0360.
    1. Dummer R, Eichmuller S, Gellrich S, Assaf C, Dreno B, Schiller M, Dereure O, Baudard M, Bagot M, Khammari A. et al.Phase II clinical trial of intratumoral application of TG1042 (adenovirus-interferon-gamma) in patients with advanced cutaneous T-cell lymphomas and multilesional cutaneous B-cell lymphomas. Mol Ther. 2010;11:1244–1247. doi: 10.1038/mt.2010.52.
    1. Olsen EA, Bunn PA. Interferon in the treatment of cutaneous T-cell lymphoma. Hematol Oncol Clin North Am. 1995;11:1089–1107.
    1. Vowels BR, Cassin M, Vonderheid EC, Rook AH. Aberrant cytokine production by Sezary syndrome patients: cytokine secretion pattern resembles murine Th2 cells. J Invest Dermatol. 1992;11:90–94. doi: 10.1111/1523-1747.ep12611877.
    1. Schroder K, Hertzog PJ, Ravasi T, Hume DA. Interferon-gamma: an overview of signals, mechanisms and functions. J Leukoc Biol. 2004;11:163–189.
    1. Seo N, Tokura Y, Matsumoto K, Furukawa F, Takigawa M. Tumour-specific cytotoxic T lymphocyte activity in Th2-type Sezary syndrome: its enhancement by interferon-gamma (IFN-gamma) and IL-12 and fluctuations in association with disease activity. Clin Exp Immunol. 1998;11:403–409. doi: 10.1046/j.1365-2249.1998.00599.x.
    1. Trudel S, Li Z, Dodgson C, Nanji S, Wan Y, Voralia M, Hitt M, Gauldie J, Graham FL, Stewart AK. Adenovector engineered interleukin-2 expressing autologous plasma cell vaccination after high-dose chemotherapy for multiple myeloma–a phase 1 study. Leukemia. 2001;11:846–854. doi: 10.1038/sj.leu.2402077.
    1. Ikeda H, Old LJ, Schreiber RD. The roles of IFN gamma in protection against tumor development and cancer immunoediting. Cytokine Growth Factor Rev. 2002;11:95–109. doi: 10.1016/S1359-6101(01)00038-7.
    1. Murphy KM, Ouyang W, Farrar JD, Yang J, Ranganath S, Asnagli H, Afkarian M, Murphy TL. Signaling and transcription in T helper development. Annu Rev Immunol. 2000;11:451–494. doi: 10.1146/annurev.immunol.18.1.451.

Source: PubMed

3
Abonner