Cancer Letters

Cancer Letters

Volume 393, 1 May 2017, Pages 52-59
Cancer Letters

Original Article
Intracavitary ‘T4 immunotherapy’ of malignant mesothelioma using pan-ErbB re-targeted CAR T-cells

https://doi.org/10.1016/j.canlet.2017.02.015Get rights and content

Highlights

  • Mesotheliomas express EGF receptor in 80% and ErbB4 in 50% of cases.

  • T-cells from mesothelioma patients are amenable to efficient genetic engineering.

  • ErbB-targeted patient CAR T-cells mediate potent activity against mesothelioma.

  • Tumor regression is not accompanied by toxicity.

  • Clinical evaluation of this approach is warranted.

Abstract

Malignant mesothelioma remains an incurable cancer. We demonstrated that mesotheliomas expressed EGFR (79.2%), ErbB4 (49.0%) and HER2 (6.3%), but lacked ErbB3. At least one ErbB family member was expressed in 88% of tumors. To exploit ErbB dysregulation in this disease, patient T-cells were engineered by retroviral transduction to express a panErbB-targeted chimeric antigen receptor (CAR), co-expressed with a chimeric cytokine receptor that allows interleukin (IL)-4 mediated CAR T-cell proliferation. This combination is referred to as T4 immunotherapy. T-cells from mesothelioma patients were uniformly amenable to T4 genetic modification and expansion/enrichment thereafter using IL-4. Patient-derived T4+ T-cells were activated upon contact with a panel of four mesothelioma cell lines, leading to cytotoxicity and cytokine release in all cases. Adoptive transfer of T4 immunotherapy to SCID Beige mice with an established bioluminescent LO68 mesothelioma xenograft was followed by regression or eradication of disease in all animals. Despite the established ability of T4 immunotherapy to elicit cytokine release syndrome in SCID Beige mice, therapy was very well tolerated. These findings provide a strong rationale for the clinical evaluation of intracavitary T4 immunotherapy to treat mesothelioma.

Introduction

Malignant mesothelioma is a locally aggressive tumor that is primarily attributable to inhalation of asbestos fibers [1], prompting stringent regulation of this mineral in many territories. Nonetheless, incidence continues to increase in many countries, reflecting the long latency period of disease progression [2]. Mesothelioma remains incurable with a median survival of 9–17 months [3], [4].

Immunotherapy has long been posited as a logical approach to treat mesothelioma [5]. Promising data have emerged from clinical trials of immune checkpoint blockade in this disease [6]. In parallel, there has been increasing interest in the development of adoptive immunotherapeutic approaches using genetically engineered T-cells. Chimeric antigen receptors (CARs) are bespoke fusion molecules that couple an antigen recognition element to a tailored signaling domain, thereby enabling the HLA independent retargeting of T-cell specificity [7]. Interest has been fueled by the repeated demonstration of unprecedented therapeutic efficacy when autologous CD19 targeted CAR T-cells are administered to patients with refractory acute lymphoblastic leukemia [8]. The most successful CAR configurations employ a second generation design in which CD28 or 4-1BB costimulatory modules are fused to CD3ζ [9], [10], [11]. While initial experience in patients with solid tumors has been disappointing, mesothelioma may represent a special case since tumors are amenable to regional CAR T-cell delivery to the affected body cavity. Preclinical data have provided strong support for this approach using CAR T-cells retargeted against mesothelin [12], [13] and fibroblast activation protein [14], [15]. Clinical trials employing both intra-pleural [16], [17] and systemic delivery [18] are ongoing.

An increasing concern with CAR T-cell immunotherapy is the emergence of therapeutic resistance due to target loss [19], [20]. This highlights the desirability of selecting multiple targets that contribute to disease pathogenesis and thus are subject to selective pressure for retention by tumors. In mesothelioma, the extended ErbB receptor family represents an attractive candidate to meet these requirements. Epidermal growth factor receptor (EGFR; ErbB1) is upregulated and phosphorylated in the majority of cases, compared to normal pleura [21], [22], [23], [24], [25], [26], [27]. Disappointingly however, clinical evaluation of EGFR tyrosine kinase inhibitors has proven ineffective in this disease [24], [25], while trials involving EGFR inhibitory antibodies are ongoing (www.clinicaltrials.gov, accessed February 5th, 2017). Co-expression of HER2 (ErbB2) [24], [28] has also been reported, although ErbB3 and ErbB4 have not previously been evaluated.

To redirect T-cell specificity against the ErbB family, we have engineered a second generation CAR named T1E28z. T1E28z engages 8 of 9 possible ErbB homo and heterodimers [29] and enables T-cells to elicit antitumor activity in preclinical models of ovarian, breast and head and neck cancers [29], [30], [31]. The CAR is co-expressed with a chimeric cytokine receptor named 4αβ that delivers an interleukin (IL)-2/IL-15 signal upon binding of IL-4, thereby enabling the selective enrichment of CAR T-cells during ex vivo expansion [32]. Importantly, culture of the resulting ‘T4+’ T-cells in IL-4 does not alter their differentiation and maintains type 1 polarity [32]. T4 immunotherapy is currently undergoing Phase 1 evaluation by intra-tumoral delivery in patients with head and neck cancer [33]. This approach has a further theoretical advantage in mesothelioma treatment since IL-4 is commonly found in the tumor microenvironment, providing an additional source of stimulation for T4+ T-cells [34]. Here, we tested the hypothesis that T4+ T-cells derived from patients with mesothelioma would exert meaningful antitumor activity against mesothelioma, justifying the clinical evaluation of this approach.

Section snippets

Materials and methods

Additional details are provided as an online supplement.

Expression of ErbB receptors by malignant pleural mesotheliomas

Tissue microarrays were prepared from 168 malignant pleural mesotheliomas (MPM). Histologic subtype was defined in 103 cases as epithelioid (56.7%), sarcomatoid (20.4%) or biphasic (22.8%). As expected [37], survival was significantly longer for patients with epithelioid mesotheliomas, compared to other subtypes (Fig. 1A).

Immunohistochemical analysis (Table 1 and Fig. 1B) demonstrated that most tumors expressed EGFR, while ErbB4 was detected in half of cases. By contrast, HER2 expression was

Discussion

Solid tumors impose considerable obstacles to effective CAR T-cell immunotherapy. The first hurdle is the delivery of sufficient numbers of T-cells to the site of disease. While disseminated metastasis represents the main therapeutic challenge imposed by most cancers, some tumors primarily advance by loco-regional dissemination. This scenario provides a unique opportunity for CAR T-cell immunotherapy since direct administration of T-cells to such lesions obviates their homing requirement from

Acknowledgements

We thank Anthony Walker, Mike Garrison and other members of the CAR Mechanics group for useful discussions. We thank Roseanna Petrovic for flow cytometry data.

This work was supported by the June Hancock Mesothelioma Research Fund [grant number JHMRF 2014], the British Lung Foundation [grant number APHD12-11], the Experimental Cancer Medicine Centre at King's College London and by the National Institute for Health Research (NIHR) Biomedical Research Centre based at Guy's and St Thomas' NHS

References (44)

  • D. Gomez et al.

    Local and systemic therapies for malignant pleural mesothelioma

    Curr. Treat. Options Oncol.

    (2014)
  • R. Cornelissen et al.

    New roads open up for implementing immunotherapy in mesothelioma

    Clin. Dev. Immunol.

    (2012)
  • G.L. Ceresoli et al.

    Immune checkpoint inhibitors in malignant pleural mesothelioma: promises and challenges

    Expert Rev. Anticancer Ther.

    (2016)
  • J. Maher

    Clinical immunotherapy of B-cell malignancy using CD19-targeted CAR T-cells

    Curr. Gene Ther.

    (2014)
  • R.J. Brentjens et al.

    CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia

    Sci. Transl. Med.

    (2013)
  • S.L. Maude et al.

    Chimeric antigen receptor T cells for sustained remissions in leukemia

    N. Engl. J. Med.

    (2014)
  • P.S. Adusumilli et al.

    Regional delivery of mesothelin-targeted CAR T cell therapy generates potent and long-lasting CD4-dependent tumor immunity

    Sci. Transl. Med.

    (2014)
  • L. Cherkassky et al.

    T cells with cell-intrinsic PD-1 checkpoint blockade resist tumor-mediated inhibition

    J. Clin. Investig.

    (2016)
  • P.C. Schuberth et al.

    Treatment of malignant pleural mesothelioma by fibroblast activation protein-specific re-directed T cells

    J. Transl. Med.

    (2013)
  • E.K. Moon et al.

    Multifactorial T-cell hypofunction that is reversible can limit the efficacy of chimeric antigen receptor-transduced human T cells in solid tumors

    Clin. Cancer Res.

    (2014)
  • M. Mayor et al.

    A regional approach for CAR T-cell therapy for mesothelioma: from mouse models to clinical trial

    Immunotherapy

    (2016)
  • M. O'Hara et al.

    Mesothelin as a target for chimeric antigen receptor-modified T cells as anticancer therapy

    Immunotherapy

    (2016)
  • Cited by (48)

    • Immunotherapy for mesothelioma: Moving beyond single immune check point inhibition

      2022, Lung Cancer
      Citation Excerpt :

      Combining this therapy with ICI may circumvent the issue of T-cell exhaustion limiting efficacy of CAR T-cells in solid malignancies like MPM [94]. While several CAR T-cell targets have been proposed (e.g., ErbB2, 5 T4, CSPG4) [56,95–101], the majority of clinical trials have focused on targeting mesothelin. Adusumilli and colleagues administered intrapleural mesothelin-targeted CAR T-cells in addition to the anti-PD1 agent pembrolizumab to 18 patients with MPM; this resulted in a median OS of 23.9 months, with 8 participants achieving disease stability for over 6 months and 2 participants a complete response on positron emission tomography (PET) imaging [102].

    • CAR T-cell therapy for pleural mesothelioma: Rationale, preclinical development, and clinical trials

      2021, Lung Cancer
      Citation Excerpt :

      In a study of MPM histologic samples, at least one ErbB family receptor was expressed on 88% of tumors, prompting the investigation of a CAR T-cell construct directed at multiple ErbB family receptors. This CAR T-cell construct inhibited tumor growth in vivo [54]. 5T4 has also been proposed as a target for CAR T-cell therapy for mesothelioma on the basis of its expression in multiple cancer types, including MPM [55], and its low expression on normal adult tissue [56].

    • Hypoxia-sensing CAR T cells provide safety and efficacy in treating solid tumors

      2021, Cell Reports Medicine
      Citation Excerpt :

      To investigate the issue of on-target, off-tumor CAR T cell activation, we utilized a second-generation pan-anti-ErbB CAR T1E28z, T4-CAR, which has specificity toward 8 of 9 of the possible ErbB receptor homo- and heterodimers and crosses the species barrier, binding human and mouse receptors equivalently.6 T4-CAR co-expresses a chimeric cytokine receptor (4αβ) that delivers an intracellular interleukin-2 (IL-2)/IL-15 signal upon binding of IL-4 to the extracellular domain, providing a means to selectively enrich CAR T cells during ex vivo expansion (Figures 1A and 1B)7–9 without affecting the CAR-dependent killing capabilities of these cells.7 T4-CAR has been proven to be safe for intra-tumoral (i.t.) delivery in individuals with head and neck squamous cell carcinoma (HNSCC),10 but i.v. infusion is required to treat individuals with metastatic disease.

    • CAR-T in solid tumors: Blazing a new trail through the brambles

      2020, Life Sciences
      Citation Excerpt :

      Similarly, Murad et al. demonstrated that the regional intraperitoneal injection of tumor-associated glycoprotein 72 (TAG72)-BBζ CAR-T cells notably inhibited tumor growth in a xenograft model of peritoneal ovarian tumors [19]. Meanwhile, the local injection of pan-ErbB/IL-4 CAR-T cells into malignant peritoneal mesothelioma xenograft mouse models by Klampatsa et al. [20] and the intraperitoneal delivery of T1E28z CAR-T cells, constructed by a promiscuous ErbB ligand fused to a CD28+CD3ζ endodomain, into intraperitoneally-implanted head and neck cancer mouse models by Davies et al. [21] showed effective anti-tumor effects. Additionally, it is well known that the central nervous system, due to the existence of the blood-brain barrier, belongs to the immune exemption site.

    View all citing articles on Scopus
    View full text