Skip to main content
Top
Published in: Cancer Immunology, Immunotherapy 8/2015

01-08-2015 | Original Article

Intralesional administration of L19-IL2/L19-TNF in stage III or stage IVM1a melanoma patients: results of a phase II study

Authors: Riccardo Danielli, Roberto Patuzzo, Anna Maria Di Giacomo, Gianfranco Gallino, Andrea Maurichi, Annabella Di Florio, Ornella Cutaia, Andrea Lazzeri, Carolina Fazio, Clelia Miracco, Leonardo Giovannoni, Giuliano Elia, Dario Neri, Michele Maio, Mario Santinami

Published in: Cancer Immunology, Immunotherapy | Issue 8/2015

Login to get access

Abstract

The intratumoral injection of cytokines, in particular IL2, has shown promise for cutaneous melanoma patients with unresectable disease or continuous recurrence despite surgery. We recently reported that the intralesional injection of L19-IL2, an immunocytokine combining IL2 and the human monoclonal antibody fragment L19, resulted in efficient regional control of disease progression, increased time to distant metastasis and evidence of effect on circulating immune cell populations. We have also shown in preclinical models of cancer a remarkable synergistic effect of the combination of L19-IL2 with L19-TNF, a second clinical-stage immunocytokine, based on the same L19 antibody fused to TNF. Here, we describe the results of a phase II clinical trial based on the intralesional administration of L19-IL2 and L19-TNF in patients with stage IIIC and IVM1a metastatic melanoma, who were not candidate to surgery. In 20 efficacy-evaluable patients, 32 melanoma lesions exhibited complete responses upon intralesional administration of the two products, with mild side effects mainly limited to injection site reactions. Importantly, we observed complete responses in 7/13 (53.8 %) non-injected lesions (4 cutaneous, 3 lymph nodes), indicating a systemic activity of the intralesional immunostimulatory treatment. The intralesional administration of L19-IL2 and L19-TNF represents a simple and effective method for the local control of inoperable melanoma lesions, with a potential to eradicate them or make them suitable for a facile surgical removal of the residual mass.
Appendix
Available only for authorised users
Literature
1.
go back to reference Testori A, Faries MB, Thompson JF, Pennacchioli E, Deroose JP, van Geel AN, Verhoef C, Verrecchia F, Soteldo J (2011) Local and intralesional therapy of in-transit melanoma metastases. J Surg Oncol 104(4):391–396. doi:10.1002/jso.22029 PubMedCrossRef Testori A, Faries MB, Thompson JF, Pennacchioli E, Deroose JP, van Geel AN, Verhoef C, Verrecchia F, Soteldo J (2011) Local and intralesional therapy of in-transit melanoma metastases. J Surg Oncol 104(4):391–396. doi:10.​1002/​jso.​22029 PubMedCrossRef
2.
go back to reference Si Z, Hersey P, Coates AS (1996) Clinical responses and lymphoid infiltrates in metastatic melanoma following treatment with intralesional GM-CSF. Melanoma Res 6(3):247–255PubMedCrossRef Si Z, Hersey P, Coates AS (1996) Clinical responses and lymphoid infiltrates in metastatic melanoma following treatment with intralesional GM-CSF. Melanoma Res 6(3):247–255PubMedCrossRef
3.
go back to reference von Wussow P, Block B, Hartmann F, Deicher H (1988) Intralesional interferon-alpha therapy in advanced malignant melanoma. Cancer 61(6):1071–1074CrossRef von Wussow P, Block B, Hartmann F, Deicher H (1988) Intralesional interferon-alpha therapy in advanced malignant melanoma. Cancer 61(6):1071–1074CrossRef
4.
go back to reference Kubo H, Ashida A, Matsumoto K, Kageshita T, Yamamoto A, Saida T (2008) Interferon-beta therapy for malignant melanoma: the dose is crucial for inhibition of proliferation and induction of apoptosis of melanoma cells. Arch Dermatol Res 300(6):297–301. doi:10.1007/s00403-008-0841-6 PubMedCrossRef Kubo H, Ashida A, Matsumoto K, Kageshita T, Yamamoto A, Saida T (2008) Interferon-beta therapy for malignant melanoma: the dose is crucial for inhibition of proliferation and induction of apoptosis of melanoma cells. Arch Dermatol Res 300(6):297–301. doi:10.​1007/​s00403-008-0841-6 PubMedCrossRef
5.
go back to reference Gutwald JG, Groth W, Mahrle G (1994) Peritumoral injections of interleukin 2 induce tumour regression in metastatic malignant melanoma. Br J Dermatol 130(4):541–542PubMedCrossRef Gutwald JG, Groth W, Mahrle G (1994) Peritumoral injections of interleukin 2 induce tumour regression in metastatic malignant melanoma. Br J Dermatol 130(4):541–542PubMedCrossRef
7.
go back to reference Weide B, Derhovanessian E, Pflugfelder A, Eigentler TK, Radny P, Zelba H, Pfohler C, Pawelec G, Garbe C (2010) High response rate after intratumoral treatment with interleukin-2: results from a phase 2 study in 51 patients with metastasized melanoma. Cancer 116(17):4139–4146. doi:10.1002/cncr.25156 PubMedCrossRef Weide B, Derhovanessian E, Pflugfelder A, Eigentler TK, Radny P, Zelba H, Pfohler C, Pawelec G, Garbe C (2010) High response rate after intratumoral treatment with interleukin-2: results from a phase 2 study in 51 patients with metastasized melanoma. Cancer 116(17):4139–4146. doi:10.​1002/​cncr.​25156 PubMedCrossRef
9.
go back to reference Dehesa LA, Vilar-Alejo J, Valeron-Almazan P, Carretero G (2009) Experience in the treatment of cutaneous in-transit melanoma metastases and satellitosis with intralesional interleukin-2. Actas Dermosifiliogr 100(7):571–585PubMedCrossRef Dehesa LA, Vilar-Alejo J, Valeron-Almazan P, Carretero G (2009) Experience in the treatment of cutaneous in-transit melanoma metastases and satellitosis with intralesional interleukin-2. Actas Dermosifiliogr 100(7):571–585PubMedCrossRef
10.
go back to reference Weide B, Eigentler TK, Pflugfelder A et al (2011) Survival after intratumoral interleukin-2 treatment of 72 melanoma patients and response upon the first chemotherapy during follow-up. Cancer Immunol Immunother 60(4):487–493. doi:10.1007/s00262-010-0957-3 PubMedCrossRef Weide B, Eigentler TK, Pflugfelder A et al (2011) Survival after intratumoral interleukin-2 treatment of 72 melanoma patients and response upon the first chemotherapy during follow-up. Cancer Immunol Immunother 60(4):487–493. doi:10.​1007/​s00262-010-0957-3 PubMedCrossRef
11.
go back to reference Weide B, Eigentler TK, Pflugfelder A et al (2014) Intralesional treatment of stage III metastatic melanoma patients with L19-IL2 results in sustained clinical and systemic immunologic responses. Cancer Immunol Res 2(7):668–678. doi:10.1158/2326-6066.CIR-13-0206 PubMedCrossRef Weide B, Eigentler TK, Pflugfelder A et al (2014) Intralesional treatment of stage III metastatic melanoma patients with L19-IL2 results in sustained clinical and systemic immunologic responses. Cancer Immunol Res 2(7):668–678. doi:10.​1158/​2326-6066.​CIR-13-0206 PubMedCrossRef
14.
go back to reference Pretto F, Elia G, Castioni N, Neri D (2014) Preclinical evaluation of IL2-based immunocytokines supports their use in combination with dacarbazine, paclitaxel and TNF-based immunotherapy. Cancer Immunol Immunother 63(9):901–910. doi:10.1007/s00262-014-1562-7 PubMedCrossRef Pretto F, Elia G, Castioni N, Neri D (2014) Preclinical evaluation of IL2-based immunocytokines supports their use in combination with dacarbazine, paclitaxel and TNF-based immunotherapy. Cancer Immunol Immunother 63(9):901–910. doi:10.​1007/​s00262-014-1562-7 PubMedCrossRef
15.
16.
go back to reference Roberts NJ, Zhou S, Diaz LA Jr, Holdhoff M (2011) Systemic use of tumor necrosis factor alpha as an anticancer agent. Oncotarget 2(10):739–751PubMedCentralPubMed Roberts NJ, Zhou S, Diaz LA Jr, Holdhoff M (2011) Systemic use of tumor necrosis factor alpha as an anticancer agent. Oncotarget 2(10):739–751PubMedCentralPubMed
17.
go back to reference Eigentler TK, Weide B, de Braud F et al (2011) A dose-escalation and signal-generating study of the immunocytokine L19-IL2 in combination with dacarbazine for the therapy of patients with metastatic melanoma. Clin Cancer Res 17(24):7732–7742. doi:10.1158/1078-0432.CCR-11-1203 PubMedCrossRef Eigentler TK, Weide B, de Braud F et al (2011) A dose-escalation and signal-generating study of the immunocytokine L19-IL2 in combination with dacarbazine for the therapy of patients with metastatic melanoma. Clin Cancer Res 17(24):7732–7742. doi:10.​1158/​1078-0432.​CCR-11-1203 PubMedCrossRef
18.
go back to reference Papadia F, Basso V, Patuzzo R et al (2013) Isolated limb perfusion with the tumor-targeting human monoclonal antibody-cytokine fusion protein L19-TNF plus melphalan and mild hyperthermia in patients with locally advanced extremity melanoma. J Surg Oncol 107(2):173–179. doi:10.1002/jso.23168 PubMedCrossRef Papadia F, Basso V, Patuzzo R et al (2013) Isolated limb perfusion with the tumor-targeting human monoclonal antibody-cytokine fusion protein L19-TNF plus melphalan and mild hyperthermia in patients with locally advanced extremity melanoma. J Surg Oncol 107(2):173–179. doi:10.​1002/​jso.​23168 PubMedCrossRef
21.
go back to reference Hassan S, Petrella T, Zhang T et al (2014) Pathologic complete response to intralesional interleukin-2 therapy associated with improved survival in melanoma patients with in-transit disease. Ann Surg Oncol. doi:10.1245/s10434-014-4199-z Hassan S, Petrella T, Zhang T et al (2014) Pathologic complete response to intralesional interleukin-2 therapy associated with improved survival in melanoma patients with in-transit disease. Ann Surg Oncol. doi:10.​1245/​s10434-014-4199-z
23.
go back to reference Kaufman HL, Kim DW, DeRaffele G, Mitcham J, Coffin RS, Kim-Schulze S (2010) Local and distant immunity induced by intralesional vaccination with an oncolytic herpes virus encoding GM-CSF in patients with stage IIIc and IV melanoma. Ann Surg Oncol 17(3):718–730. doi:10.1245/s10434-009-0809-6 PubMedCrossRef Kaufman HL, Kim DW, DeRaffele G, Mitcham J, Coffin RS, Kim-Schulze S (2010) Local and distant immunity induced by intralesional vaccination with an oncolytic herpes virus encoding GM-CSF in patients with stage IIIc and IV melanoma. Ann Surg Oncol 17(3):718–730. doi:10.​1245/​s10434-009-0809-6 PubMedCrossRef
24.
go back to reference Lotze MT (1995) Biologic therapy with interleukin-2: preclinical studies. In: DeVita VTJ, Hellman S, Rosenberg SA (eds) Biologic therapy of cancer. Lippincott, Philadelphia, pp 207–233 Lotze MT (1995) Biologic therapy with interleukin-2: preclinical studies. In: DeVita VTJ, Hellman S, Rosenberg SA (eds) Biologic therapy of cancer. Lippincott, Philadelphia, pp 207–233
27.
go back to reference Berntsen A, Brimnes MK, thor Straten P, Svane IM (2010) Increase of circulating CD4+ CD25highFoxp3+ regulatory T cells in patients with metastatic renal cell carcinoma during treatment with dendritic cell vaccination and low-dose interleukin-2. J Immunother 33(4):425–434. doi:10.1097/CJI.0b013e3181cd870f PubMedCrossRef Berntsen A, Brimnes MK, thor Straten P, Svane IM (2010) Increase of circulating CD4+ CD25highFoxp3+ regulatory T cells in patients with metastatic renal cell carcinoma during treatment with dendritic cell vaccination and low-dose interleukin-2. J Immunother 33(4):425–434. doi:10.​1097/​CJI.​0b013e3181cd870f​ PubMedCrossRef
28.
go back to reference Martens A, Zelba H, Garbe C, Pawelec G, Weide B (2014) Monocytic myeloid-derived suppressor cells in advanced melanoma patients: indirect impact on prognosis through inhibition of tumor-specific T-cell responses? Oncoimmunology 3(1):e27845. doi:10.4161/onci.27845 PubMedCentralPubMedCrossRef Martens A, Zelba H, Garbe C, Pawelec G, Weide B (2014) Monocytic myeloid-derived suppressor cells in advanced melanoma patients: indirect impact on prognosis through inhibition of tumor-specific T-cell responses? Oncoimmunology 3(1):e27845. doi:10.​4161/​onci.​27845 PubMedCentralPubMedCrossRef
29.
go back to reference Weide B, Martens A, Zelba H et al (2014) Myeloid-derived suppressor cells predict survival of patients with advanced melanoma: comparison with regulatory T cells and NY-ESO-1- or melan-A-specific T cells. Clin Cancer Res 20(6):1601–1609. doi:10.1158/1078-0432.CCR-13-2508 PubMedCrossRef Weide B, Martens A, Zelba H et al (2014) Myeloid-derived suppressor cells predict survival of patients with advanced melanoma: comparison with regulatory T cells and NY-ESO-1- or melan-A-specific T cells. Clin Cancer Res 20(6):1601–1609. doi:10.​1158/​1078-0432.​CCR-13-2508 PubMedCrossRef
30.
go back to reference Fadaki N, Cardona-Huerta S, Martineau L et al (2012) Inoperable bulky melanoma responds to neoadjuvant therapy with vemurafenib. BMJ Case Rep 2012. doi:10.1136/bcr-2012-007034 Fadaki N, Cardona-Huerta S, Martineau L et al (2012) Inoperable bulky melanoma responds to neoadjuvant therapy with vemurafenib. BMJ Case Rep 2012. doi:10.​1136/​bcr-2012-007034
Metadata
Title
Intralesional administration of L19-IL2/L19-TNF in stage III or stage IVM1a melanoma patients: results of a phase II study
Authors
Riccardo Danielli
Roberto Patuzzo
Anna Maria Di Giacomo
Gianfranco Gallino
Andrea Maurichi
Annabella Di Florio
Ornella Cutaia
Andrea Lazzeri
Carolina Fazio
Clelia Miracco
Leonardo Giovannoni
Giuliano Elia
Dario Neri
Michele Maio
Mario Santinami
Publication date
01-08-2015
Publisher
Springer Berlin Heidelberg
Published in
Cancer Immunology, Immunotherapy / Issue 8/2015
Print ISSN: 0340-7004
Electronic ISSN: 1432-0851
DOI
https://doi.org/10.1007/s00262-015-1704-6

Other articles of this Issue 8/2015

Cancer Immunology, Immunotherapy 8/2015 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine