Potential dual synergy between electrochemotherapy and sequence of immunotherapies in metastatic melanoma: A case report

  • Authors:
    • Francesca Morgese
    • Francesco De Feudis
    • Paolo Balercia
    • Rossana Berardi
  • View Affiliations

  • Published online on: January 10, 2023     https://doi.org/10.3892/mco.2023.2604
  • Article Number: 8
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Immune checkpoint inhibitors have changed the natural history of advanced melanoma. Despite this, a notable proportion of patients immediately relapse or develop resistance during immunotherapy, especially with the appearance of superficial metastases and consequently with a dramatic impact on clinical outcomes. Local treatment by electrochemotherapy (ECT), parallel to regional control with palliative aim, seems to release neoantigens potentially determining a significant systemic anticancer immune reactivation. The present study reported a case of a patient with metastatic melanoma receiving Pembrolizumab, electrochemotherapy and then Ipilimumab for in‑transit and finally locoregional lymph nodes and distant bone metastases with experience of clinic‑radiological remission. Specifically, the present patient progressed during adjuvant treatment with in‑transit metastases on the scalp; he underwent two cycle of ECT obtaining partial and then unexpected and very fast nearly complete response with the Ipilimumab treatment. Concomitantly, he developed grade 4 endocrine adverse events (hypophysitis and diabetes mellitus type I) as immune‑related toxicities. At 12 months from ECT the patient is in ECOG Performance Status 0 and he has resumed a regular social life. In our experience, ECT in two administrations increased and accelerated the response of Ipilimumab. The present confirmed its promising contribution in inducing a powerful immune response in order to overcome primary or acquired resistance to immune checkpoint inhibitors such as anti‑programmed death antigen‑1 drugs.

Introduction

Recently, novel immunotherapies directed against CTLA-4 (Cytotoxic T Lymphocyte Antigen-4) and PD-1 (Programmed Death antigen-1) afford the opportunity to effectively treat loco-regional and metastatic melanoma, ensuring significant reduction of risk for relapse, with relatively durable responses and definitely with a considerable improvement of survival (1,2).

Monotherapy with anti-PD-1 drugs, i.e. Nivolumab or Pembrolizumab, represents the standard adjuvant treatment in BRAF wild type melanoma patients (3,4).

The combination of Nivolumab and anti-CTLA4, Ipilimumab, in BRAF wild type and negative PD-L1 tumors, or anti-PD1 alone in melanoma with PD-L1 expression, are instead the most recent two options for metastatic disease (1,5).

Nevertheless, in this new therapeutic scenario, over half patients with melanoma, especially melanotic one, during or immediately after adjuvant anti-PD1 treatment recurs, or does not respond to modern immunotherapy or, moreover, relapses after a temporary response (1,3-5).

For patients with primary resistance or progressing to anti-PD-1 or even to the combination the prognosis is extremely unfavorable, with a second/following line including Ipilimumab, alkylating chemotherapy, or agents in experimental trials.

Among them, there are many patients that can develop peculiar refractory superficial metastases.

In-transit metastases represent cutaneous localization of melanoma that appear between a primary tumor and its regional lymph node basin as part of the natural lymphatic dissemination process. In presence of lymph node involvement their incidence is 20% (6).

The prognosis of patients with in-transit melanoma metastases is unfavorable with 5-year survival ranging from 12 to 37% (7).

The surgical excision of isolated lesions represents the elective treatment.

For metastases, which primarily show or develop resistance to standard therapies, electroporation associated with chemotherapy (electrochemotherapy-ECT) seems to be a new potential therapeutic option in particular in combination with immunotherapy, since it elicits a systemic immune response other than directly treat deep-seated tumors. The biological rational is represented by the ECT induction of immunogenic cancer cells death. In fact, this procedure can determine a more efficient antigen presentation of tumor-derived antigens by APCs (antigen-presenting cells), particularly CD8+ antigen-specific T cells. Anti-PD-1 drugs revert T-cell exhaustion induced by PD-1/PD-L1 (programmed death-ligand 1) and PD-L2 (programmed death-ligand 2) engagement on CD8+ T cells. Moreover, ECT may also be beneficial in the priming phase of the antitumor immune responses, provoking long-lived tumor antigen-specific CD8+ T-cell effectors (8).

However no data from trials exist to evaluate the potential role of the combination or the correct sequence of these two therapeutic options-loco-regional and systemic treatment-but only anecdotic/retrospective cases in papers. Moreover, nowadays it is unknown the appropriate timing and sequence in terms of restoring efficacy of immunotherapy in resistance events (9-13).

Our experience on a patient with initial in-transit metastases on the scalp and then loco regional lymph node and bone metastases showed the crucial beneficial role of a peculiar sequence of electrochemotherapy and immunotherapies.

Case report

A 73-year old male patient with no significant medical history, except for rheumatic polymyalgia regressed after steroid therapy, was subjected to excision of cutaneous lesion on the scalp in November 2020. The histological exam revealed a lentigo-maligna melanoma in vertical growth phase, with the following features: Clark level IV invasion, 1.7 mm Breslow index, 10 mitoses/mm2, TIL non brisk, absence of ulceration, regression, microsatellitosis and lymphovascular invasion, presence of perineural invasion and of melanin pigmentation.

In December 2020, the patient underwent intra-parotid sentinel lymph nodes biopsy (SLNB) and radicalization of previous exeresis. The histological skin-related report was positive for the presence of multiple melanoma foci accountable as satellite nodules of lentigo-maligna melanoma infiltrating the dermis for a maximum thickness of 1.1 mm (greater nodule). The parenchyma of salivary gland resulted free from pathological lesions. The histological examination of SLNB in the intra and periglandular tissue showed: one of these, in its subcapsular location, was positive for rare cells ascribable to metastases from melanoma. Total body CT-scan resulted in plausible no evidence of loco-regional and distant metastases (some unspecific micro-nodular to pulmonary parenchyma bilaterally, mainly to the upper lung lobes and angiomatous lesion of ~68 mm localized in the fourth hepatic segment, known in patient's history).

Furthermore, no BRAF gene mutation was identified. It ended for an onset stage IIIC (AJCC 2017) of the melanoma disease (pT2apN2cM0).

In January 2021 the patient started anti-PD1 Pembrolizumab 200 mg flat dose therapy with adjuvant purpose.

For the appearance of a suspected pigmented peri-scar skin lesion on the scalp (frontal region), the patient underwent surgery in May 2021. Histological examination confirmed the clinical suspicion of in-transit metastases.

A total body CT-scan of June 2021 showed unchanged findings and the patient continued Pembrolizumab without interruptions.

Subsequently for the rapidly progressive disease on the scalp in May-June 2021, the patient, after discussion in the multidisciplinary context and signing the informed consent to the treatment, was subjected to ECT to treat the scalp and its superficial lymphatic drainage pathways. Considering the oligo-progression of the disease (no visceral and brain melanoma localizations), the characteristics resistance of cutaneous lesions to elective treatments and the potential synergy between ECT and immunotherapy, the patient followed the same oncological treatment as first line therapy.

In particular, he underwent two ECT administrations with intravenous injection of bleomycin 30,000 UI through linear electrode and with a 6 weeks interval (June 2021-131 pulses- and August 2021-273 pulses-). He showed a partial response (persistence of disease on the left frontal-temporal region) and no significant adverse events [only pruritus G2 according to common toxicities criteria adverse events (CTCAE) version 5.0 was referred] Fig. 1, Fig. 2 and Fig. 3.

In September 2021, the patient repeated a total body CT-scan with the evidence of a strange behavior of the known liver lesion (dimensional reduction from 68 to 40 mm) Fig. 4.

To better define the subsequent therapeutic process, the multidisciplinary team decided for a diagnostic investigation using 18-F-FDG PET-CT scan.

PET-CT scan carried out in October 2021 showed pathological uptake in the right iliac bone, in the right intra-parotid and left preauricular lymph nodes, and in the in-transit metastases especially at the left frontal-parietal region. There was no liver uptake. Thus, the case was discussed with radiologists: the focal lesion of liver seemed to be a fibrotic involution of the known angioma, but the appearance of lymph nodes in the neck could indicate a progression of disease. Instead, for the complex identification of intramedullary localization by CT scan, the time of onset of bone metastasis remained uncertain Fig. 5.

No clinical trials were active for our patient, thus he started a second line therapy with anti-CTLA4 Ipilimumab. After the first administration the patient had immediately benefit with the resolution of feeling of encumbrance of lymph node in parotid region and with the remission of the known nodules of the scalp. Nevertheless after four canonical doses of Ipilimumab ended in February 2022, the patient developed acute hypophysitis (grade 4-G4 according to CTCAE version 5.0). This peculiar immune-related adverse event was characterized by asthenia, hyporexia with consequent dehydration and hospital admission. He performed RMN of the sella turcica with reduction of physiological contrast uptake in the neurohypophysis. Thus a replacement steroid therapy was started after the load dose because of the corticotropic axis failure, with immediate advantage.

Then, he was hospitalized again for evidence of significant asymptomatic hyperglycemia (G4 CTCAE 5.0). Thus, diabetes mellitus type I was diagnosed and the patient undertook insulin therapy.

PET-CT scan performed in March 2022 showed a complete response of disease Fig. 6.

During Pembrolizumab treatment we observed the onset of lymphopenia; instead eosinophilia, progressive lymphocyte increase to normal values and lastly neutropenia appeared firstly during the ECT and then with Ipilimumab.

Although these adverse effects, the patient had a very fast and considerable improvement on its quality of life especially for the remission of strongly pronounced skin-lesions on the scalp. Until now, after 12 months from the first ECT and 6 months from the starting of anti-CTLA4 immunotherapy, the patient is in excellent clinical condition and complete response of disease still maintains Fig. 7.

Discussion

In this manuscript we reported a case report of metastatic melanotic melanoma patient receiving Pembrolizumab, electrochemotherapy and then Ipilimumab for cutaneous and finally loco-regional lymph nodes and distant bone metastases with experience of dramatic clinical-radiological benefit.

Specifically our patient progressed during adjuvant treatment with in-transit melanotic metastases on the scalp. It is reported that melanin pigment and melanogenesis have a crucial function in the progression of melanotic melanoma determining resistance to immunotherapy, probably by glycolysis and hypoxia-inducible factor 1-alpha (HIF-1a) activation and their immunosuppressive effects. In fact a negative correlation between tumor pigmentation and diseases outcome was shown (14).

Therefore, after the discussion in the Skin Cancer Multidisciplinary Group of our University Hospital, to overcome the melanotic metastasis' resistance to Pembrolizumab, the patient underwent electrochemotherapy obtaining partial and then nearly complete response with the Ipilimumab treatment.

The electrochemotherapy was safe; the patient only reported pruritus in the scalp, treated with antihistamine medications. During the treatment with Ipilimumab the patient developed hypophysitis with corticotropic axis failure and type 1 diabetes mellitus: thus an insulin therapy and cortisone acetate was started.

ECT combines the inducted electroporation of cancer cells with the concurrent infusion of bleomycin as cytotoxic chemotherapy, more often than cisplatin. A pulsed electrical current enhances the cell permeability expounding the event of revocable electroporation. ECT also determines anti-vascular effect resulting in increased tumor cell hypoxia. Therefore, locally bleomycin is more active with negligible systemic adverse events (15,16).

In addition to determine cell death, ECT can generate a local and systemic immune reaction as a result of releasing tumor associated antigens (TAA) from electroporated cancer cells.

TAA are neoantigens able to evoke an immune response primarily mediated by cytotoxic-T lymphocytes (CTL). Furthermore, TAA seem to be captured by local dendritic cells and then presented to tumor-specific CTL in draining lymph nodes (17-20).

Some reports showed the efficacy of ECT in the treatment of in-transit or subcutaneous metastases from cutaneous melanoma. An objective response rate from 60 to 90% was achieved in the palliative management of unresectable recurrent cutaneous disease. This response is usually long-lasting; moreover ECT is an easy, rapid and effective procedure which can be repeated (21-23). However, a long-term complete response of these peculiar localizations of metastasis seems to be difficult to obtain due to spread of cancer cells into lymphatic vessels (24-27).

In a similar way as ECT, a systemic and unexpected response to a local treatment derives from the radiotherapy of lesions, due to the release of TAA, named abscopal effect. It has been also demonstrated in patients affected by metastatic melanoma: distant lesions have showed a clinical-radiological response.

Thus, it has been hypothesized that the association of ECT with anti-PD1/anti-CTLA4 could represent an effective strategy to induce an immunological durable and synergic response against the cancer (11,28-32).

The role of hypoxia in this combination treatment has virtually been examined, particularly in melanotic melanoma. The response to radiation/ECT joint with immune checkpoint inhibition seems to be dependent on the hypoxia level (33) that could be measured to prescribe appropriate dose of radiotherapy and the number of ECT administrations.

Trials utilizing concomitant stereotactic body radiotherapy/and immunotherapy are still ongoing or recently completed without results about their synergistic impact and the best schedule for the two treatments (NCT02659540 with preliminary results¸ NCT04581382, NCT03850691, NCT03297463, NCT02406183, NCT04017897). Only one trial-NCT03448666-, still ongoing, aims to evaluate the activity of ECT and Pembrolizumab in patients with superficial or superficial and visceral metastases. ECT is administrated after the first cycle of immunotherapy. Starting with ECT could determine intralesional necrosis reducing loco-regional immunotherapy efficacy; instead, ECT administration after the first dispensation demonstrated a synergistic effect in retrospective series (11), probably due to the aforementioned mechanism of neo-antigens release. However, immunotherapy alone could induce an optimal local and systemic performance without the use of ECT. Thus, the immunotherapy-ECT sequence can be considered in oligo-progressing disease, for instance.

Considering that Ipilimumab shows uncommon, not immediate but long lasting action: usually the complete response was achieved after some months from the start (34). According to our experience, ECT leads to a local response and probably to an acceleration of the systemic one. Thus, we hypothesize that ECT in two administrations could increase and accelerate the efficacy of Ipilimumab.

Finally, randomized clinical trials and translational researches are needed to shed the light on the improved combination/sequence/number of administrations of ECT and systemic immunotherapy in order to offer the most appropriate therapies, especially for patients with superficial (and also visceral) metastases.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

FM and FDF performed the patient treatments (immunotherapy and electrochemotherapy respectively) and they were major contributors in writing the paper and contributed to design and conception. They verified efficacy, monitored patient and collected images and medical data. RB and PB formulated and supervised treatment plans. FM and FDF confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The patient consented to the collection of data and images for the aim of research and for publication in written form.

Patient consent for publication

Written consent was obtained from the patient. He authorized us to publish his disease history and his images.

Competing interests

FM was a consultant/advisory board member for BMS and MSD. The rest of the authors declare that they have no competing interests.

References

1 

Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, et al: Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 363:711–723. 2010.PubMed/NCBI View Article : Google Scholar

2 

Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Rutkowski P, Lao CD, Cowey CL, Schadendorf D, Wagstaff J, Dummer R, et al: Five-year survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med. 381:1535–1546. 2019.PubMed/NCBI View Article : Google Scholar

3 

Weber J, Mandala M, Del Vecchio M, Gogas HJ, Arance AM, Cowey CL, Dalle S, Schenker M, Chiarion-Sileni V, Marquez-Rodas I, et al: Adjuvant nivolumab versus ipilimumab in resected stage III or IV melanoma. N Engl J Med. 377:1824–1835. 2017.PubMed/NCBI View Article : Google Scholar

4 

Eggermont AMM, Blank CU, Mandalà M, Long GV, Atkinson VG, Dalle S, Haydon AM, Meshcheryakov A, Khattak A, Carlino MS, et al: Adjuvant pembrolizumab versus placebo in resected stage III melanoma (EORTC 1325-MG/KEYNOTE-054): Distant metastasis-free survival results from a double-blind, randomised, controlled, phase 3 trial. Lancet Oncol. 22:643–654. 2021.PubMed/NCBI View Article : Google Scholar

5 

Robert C, Ribas A, Schachter J, Arance A, Grob JJ, Mortier L, Daud A, Carlino MS, McNeil CM, Lotem M, et al: Pembrolizumab versus ipilimumab in advanced melanoma (KEYNOTE-006): Post-hoc 5-year results from an open-label, multicentre, randomised, controlled, phase 3 study. Lancet Oncol. 20:1239–1251. 2019.PubMed/NCBI View Article : Google Scholar

6 

Kretschmer L, Beckmann I, Thoms KM, Mitteldorf C, Bertsch HP and Neumann C: Factors predicting the risk of in-transit recurrence after sentinel lymphonodectomy in patients with cutaneous malignant melanoma. Ann Surg Oncol. 13:1105–1112. 2006.PubMed/NCBI View Article : Google Scholar

7 

Pawlik TM, Ross MI, Thompson JF, Eggermont AM and Gershenwald JE: The risk of in-transit melanoma metastasis depends on tumor biology and not the surgical approach to regional lymph nodes. J Clin Oncol. 23:4588–4590. 2005.PubMed/NCBI View Article : Google Scholar

8 

Campana LG, Testori A, Mozzillo N and Rossi CR: Treatment of metastatic melanoma with electrochemotherapy. J Surg Oncol. 109:301–307. 2014.PubMed/NCBI View Article : Google Scholar

9 

Quaresmini D, Di Lauro A, Fucci L, Strippoli S, De Risi I, Sciacovelli AM, Albano A, Achille G, Montepara M, Russo S, et al: Electrochemotherapy as a trigger to overcome primary resistance to Anti-PD-1 treatment: A case report of melanoma of the scalp. Front Oncol. 11(742666)2021.PubMed/NCBI View Article : Google Scholar

10 

Brizio M, Fava P, Astrua C, Cavaliere G and Savoia P: Complete regression of melanoma skin metastases after electrochemotherapy plus ipilimumab treatment: An unusual clinical presentation. Eur J Dermatol. 25:271–272. 2015.PubMed/NCBI View Article : Google Scholar

11 

Mozzillo N, Simeone E, Benedetto L, Curvietto M, Giannarelli D, Gentilcore G, Camerlingo R, Capone M, Madonna G, Festino L, et al: Assessing a novel immuno-oncology-based combination therapy: Ipilimumab plus electrochemotherapy. Oncoimmunology. 4(e1008842)2015.PubMed/NCBI View Article : Google Scholar

12 

Heppt MV, Eigentler TK, Kähler KC, Herbst RA, Göppner D, Gambichler T, Ulrich J, Dippel E, Loquai C, Schell B, et al: Immune checkpoint blockade with concurrent electrochemotherapy in advanced melanoma: A retrospective multicenter analysis. Cancer Immunol Immunother. 65:951–959. 2016.PubMed/NCBI View Article : Google Scholar

13 

Karaca B, Yayla G, Erdem M and Gürler T: Electrochemotherapy with anti-PD-1 treatment induced durable complete response in heavily pretreated metastatic melanoma patient. Anticancer Drugs. 29:190–196. 2018.PubMed/NCBI View Article : Google Scholar

14 

Slominski RM, Sarna T, Płonka PM, Raman C, Brożyna AA and Slominski AT: Melanoma, melanin, and melanogenesis: The Yin and Yang relationship. Front Oncol. 12(842496)2022.PubMed/NCBI View Article : Google Scholar

15 

Sersa G, Miklavcic D, Cemazar M, Rudolf Z, Pucihar G and Snoj M: Electrochemotherapy in treatment of tumours. Eur J Surg Oncol. 34:232–240. 2008.PubMed/NCBI View Article : Google Scholar

16 

Kranjc S, Kranjc M, Scancar J, Jelenc J, Sersa G and Miklavcic D: Electrochemotherapy by pulsed electromagnetic field treatment (PEMF) in mouse melanoma B16F10 in vivo. Radiol Oncol. 50:39–48. 2016.PubMed/NCBI View Article : Google Scholar

17 

Roux S, Bernat C, Al-Sakere B, Ghiringhelli F, Opolon P, Carpentier AF, Zitvogel L, Mir LM and Robert C: Tumor destruction using electrochemotherapy followed by CpG oligodeoxynucleotide injection induces distant tumor responses. Cancer Immunol Immunother. 57:1291–1300. 2008.PubMed/NCBI View Article : Google Scholar

18 

Di Gennaro P, Gerlini G, Urso C, Sestini S, Brandani P, Pimpinelli N and Borgognoni L: CD4+FOXP3+ T regulatory cells decrease and CD3+CD8+ T cells recruitment in TILs from melanoma metastases after electrochemotherapy. Clin Exp Metastasis. 33:787–798. 2016.PubMed/NCBI View Article : Google Scholar

19 

Gerlini G, Sestini S, Di Gennaro P, Urso C, Pimpinelli N and Borgognoni L: Dendritic cells recruitment in melanoma metastasis treated by electrochemotherapy. Clin Exp Metastasis. 30:37–45. 2013.PubMed/NCBI View Article : Google Scholar

20 

Liu CC, Yang H, Zhang R, Zhao JJ and Hao DJ: Tumour-associated antigens and their anti-cancer applications. Eur J Cancer Care. 26(e12446)2017.PubMed/NCBI View Article : Google Scholar

21 

Sersa G, Stabuc B, Cemazar M, Miklavcic D and Rudolf Z: Electrochemotherapy with cisplatin: The systemic antitumour effectiveness of cisplatin can be potentiated locally by the application of electric pulses in the treatment of malignant melanoma skin metastases. Melanoma Res. 10:381–385. 2000.PubMed/NCBI View Article : Google Scholar

22 

Kaehler KC, Egberts F and Hauschild A: Electrochemotherapy in symptomatic melanoma skin metastases: Intraindividual comparison with conventional surgery. Dermatol Surg. 36:1200–1202. 2010.PubMed/NCBI View Article : Google Scholar

23 

Mozzillo N, Caracò C, Mori S, Di Monta G, Botti G, Ascierto PA, Caracò C and Aloj L: Use of neoadjuvant electrochemotherapy to treat a large metastatic lesion of the cheek in a patient with melanoma. J Transl Med. 10(131)2012.PubMed/NCBI View Article : Google Scholar

24 

Möller MG, Salwa S, Soden DM and O'Sullivan GC: Electrochemotherapy as an adjunct or alternative to other treatments for unresectable or in-transit melanoma. Expert Rev Anticancer Ther. 9:1611–1630. 2009.PubMed/NCBI View Article : Google Scholar

25 

Kis E, Oláh J, Ócsai H, Baltas E, Gyulai R, Kemény L and Horvath AR: Electrochemotherapy of cutaneous metastases of melanoma-a case series study and systematic review of the evidence. Dermatol Surg. 37:816–824. 2011.PubMed/NCBI View Article : Google Scholar

26 

Colombo GL, Di Matteo S and Mir LM: Cost-effectiveness analysis of electrochemotherapy with the Cliniporator vs other methods for the control and treatments of cutaneous and subcutaneous tumors. Therap Clin Risk Manag. 4:541–548. 2008.PubMed/NCBI View Article : Google Scholar

27 

Campana LG, Mocellin S, Basso M, Puccetti O, De Salvo GL, Chiarion-Sileni V, Vecchiato A, Corti L, Rossi CR and Nitti D: Bleomycin-based electrochemotherapy: Clinical outcome from a single institution's experience with 52 patients. Ann Surg Oncol. 16:191–199. 2009.PubMed/NCBI View Article : Google Scholar

28 

Snoj M, Paulin-Kosir Z, Cemazar S and Sersa G: Long lasting complete response in melanoma treated by electrochemotherapy. Eur J Cancer. 4 (Suppl)(S2)2006.

29 

Queirolo P, Marincola F and Spagnolo F: Electrochemotherapy for the management of melanoma skin metastasis: A review of the literature and possible combinations with immunotherapy. Arch Dermatol Res. 306:521–526. 2014.PubMed/NCBI View Article : Google Scholar

30 

Goggins CA and Khachemoune A: The use of electrochemotherapy in combination with immunotherapy in the treatment of metastatic melanoma: A focused review. Int J Dermatol. 58:865–870. 2019.PubMed/NCBI View Article : Google Scholar

31 

Longo F, Perri F, Caponigro F, Della Vittoria Scarpati G, Guida A, Pavone E, Aversa C, Muto P, Giuliano M, Ionna F and Solla R: Boosting the immune response with the combination of electrochemotherapy and immunotherapy: A new weapon for squamous cell carcinoma of the head and neck? Cancers (Basel). 12(2781)2020.PubMed/NCBI View Article : Google Scholar

32 

Maglietti F, Tellado M, De Robertis M, Michinski S, Fernández J, Signori E and Marshall G: Electroporation as the immunotherapy strategy for cancer in veterinary medicine: State of the art in Latin America. Vaccines (Basel). 8(537)2020.PubMed/NCBI View Article : Google Scholar

33 

Hompland T, Fjeldbo CS and Lyng H: Tumor hypoxia as a barrier in cancer therapy: Why levels matter. Cancers (Basel). 13(499)2021.PubMed/NCBI View Article : Google Scholar

34 

Saenger YM and Wolchok JD: The heterogeneity of the kinetics of response to ipilimumab in metastatic melanoma: Patient cases. Cancer Immun. 8(1)2008.PubMed/NCBI

Related Articles

Journal Cover

February-2023
Volume 18 Issue 2

Print ISSN: 2049-9450
Online ISSN:2049-9469

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Morgese F, De Feudis F, Balercia P and Berardi R: Potential dual synergy between electrochemotherapy and sequence of immunotherapies in metastatic melanoma: A case report. Mol Clin Oncol 18: 8, 2023
APA
Morgese, F., De Feudis, F., Balercia, P., & Berardi, R. (2023). Potential dual synergy between electrochemotherapy and sequence of immunotherapies in metastatic melanoma: A case report. Molecular and Clinical Oncology, 18, 8. https://doi.org/10.3892/mco.2023.2604
MLA
Morgese, F., De Feudis, F., Balercia, P., Berardi, R."Potential dual synergy between electrochemotherapy and sequence of immunotherapies in metastatic melanoma: A case report". Molecular and Clinical Oncology 18.2 (2023): 8.
Chicago
Morgese, F., De Feudis, F., Balercia, P., Berardi, R."Potential dual synergy between electrochemotherapy and sequence of immunotherapies in metastatic melanoma: A case report". Molecular and Clinical Oncology 18, no. 2 (2023): 8. https://doi.org/10.3892/mco.2023.2604