Advances in immunotherapy for melanoma

BMC Medicine, Feb 2016

In recent years, the introduction and Federal Drug Administration approval of immune checkpoint inhibitor antibodies has dramatically improved the clinical outcomes for patients with advanced melanoma. These antagonist monoclonal antibodies are capable of unleashing dormant or exhausted antitumor immunity, which has led to durable complete and partial responses in a large number of patients. Ipilimumab targets the cytotoxic T lymphocyte-associated protein 4 (CTLA-4) receptor. Nivolumab and pembrolizumab target programmed cell death protein 1 (PD-1) receptors and have proven to be superior to ipilimumab alone. The combination of ipilimumab and nivolumab has yielded higher response rates, greater tumor shrinkage, and longer progression-free survival than either monotherapy alone. As other promising immunotherapies for melanoma proceed through clinical trials, future goals include defining the role of immune checkpoint inhibitors as adjuvant therapy, identifying optimal combination strategies, and developing reliable predictive biomarkers to guide treatment selection for individual patients.

A PDF file should load here. If you do not see its contents the file may be temporarily unavailable at the journal website or you do not have a PDF plug-in installed and enabled in your browser.

Alternatively, you can download the file locally and open with any standalone PDF reader:

http://www.biomedcentral.com/content/pdf/s12916-016-0571-0.pdf

Advances in immunotherapy for melanoma

Redman et al. BMC Medicine Advances in immunotherapy for melanoma Jason M. Redman 0 1 Geoffrey T. Gibney 0 1 Michael B. Atkins 0 1 0 Department of Medicine, Georgetown University Medical Center , Washington DC , USA 1 Georgetown Lombardi Comprehensive Cancer Center 3970 Reservoir Road, NW Research Building, Room E501, 20007 Washington DC , USA In recent years, the introduction and Federal Drug Administration approval of immune checkpoint inhibitor antibodies has dramatically improved the clinical outcomes for patients with advanced melanoma. These antagonist monoclonal antibodies are capable of unleashing dormant or exhausted antitumor immunity, which has led to durable complete and partial responses in a large number of patients. Ipilimumab targets the cytotoxic T lymphocyte-associated protein 4 (CTLA-4) receptor. Nivolumab and pembrolizumab target programmed cell death protein 1 (PD-1) receptors and have proven to be superior to ipilimumab alone. The combination of ipilimumab and nivolumab has yielded higher response rates, greater tumor shrinkage, and longer progression-free survival than either monotherapy alone. As other promising immunotherapies for melanoma proceed through clinical trials, future goals include defining the role of immune checkpoint inhibitors as adjuvant therapy, identifying optimal combination strategies, and developing reliable predictive biomarkers to guide treatment selection for individual patients. Anti-PD-1; Immunotherapy; Ipilimumab; Melanoma; Nivolumab; Pembrolizumab Background Advanced melanoma has historically been associated with a poor prognosis, with a median overall survival (OS) of 8–10 months and a 5-year survival rate of 10 % [ 1 ]. Chemotherapy clinical trials produced modest benefit to patients, with short-lived objective responses typically seen in less than 15 % of patients [ 2 ]. Initial studies in the 1980s demonstrated the ability of interleukin-2 (IL-2) to mediate tumor regression in melanoma and other malignancies [ 3 ]. In addition, it was recognized that patients with melanoma tumors infiltrated with T cells had better long-term survival, potentially as a result of an active antitumor response by the immune system, which led to therapeutic approaches using recombinant high-dose IL-2 to induce immune-mediated tumor cell lysis in patients with metastatic melanoma [ 4, 5 ]. Pooled data from patients treated at the National Cancer Institute and within the Extramural IL-2 Working Group demonstrated objective responses in 16 % of patients treated with high-dose IL-2 [6], of which, nearly half were durable or permanent, suggesting that long-term survival or ‘cure’ is possible. However, IL-2 is associated with a number of serious toxicities, largely related to vascular leak syndrome, requiring inpatient management at experienced centers. While these factors have limited its generalized use, high-dose IL-2 serves as proof of principle that immunotherapies can eliminate tumor cells in some patients, encouraging efforts to develop better tolerated and more effective immunotherapy regimens. In order to achieve antitumor effects, cytotoxic T lymphocytes (CTL) must not only migrate to the tumor, but must also be capable of tumor cell lysis. While the presence of tumor infiltrating lymphocytes (TIL) is frequently seen in melanoma tumors, TILs often have a diminished capacity for proliferation, cytokine production, and tumor lysis [ 7 ]. However, when TILs are removed from the tumor microenvironment (TME) and grown ex vivo, they can exhibit potent and specific antitumor activity, implying that the immune climate within the TME can dampen CTL activity. Evidence suggests that inflammation caused by immune infiltration can induce immune escape mechanisms, including interferon (IFN)gamma-mediated upregulation of programmed deathligand 1 (PD-L1) in the TME and increased numbers of regulatory T cells (Tregs) [ 8 ]. The engagement of PD-L1 (and PD-L2) with the programmed cell death protein 1 (PD-1) receptor on CTL leads to T cell exhaustion. Antibodies to PD-1 or PD-L1 have been shown to block the interaction between these molecules and restore antitumor immunity within the TME [ 9, 10 ]. Another mechanism of dampened immune response that is thought to predominately exert its effects in secondary lymphoid organs, as opposed to within the TME, involves cytotoxic T lymphocyte-associated protein 4 (CTLA-4) expression on T cells. CTLA-4 is a receptor exclusively expressed on T cells that binds to CD80 (B7.1) and CD86 (B7.2) on antigen-presenting cells [ 11 ]. T cell inhibition via this receptor occurs through multiple mechanisms. By outcompeting CD28 for binding to B7.1 and B7.2, CTLA-4 can prevent the co-stimulation necessary to generate and maintain T cell activation. Additionally, evidence suggests that expression of CTLA-4 on Tregs is important in T cell inhibition [ 12 ]. Antibodies to CTLA-4 have been shown to block the interaction between CTLA-4 and (...truncated)


This is a preview of a remote PDF: http://www.biomedcentral.com/content/pdf/s12916-016-0571-0.pdf

Jason Redman, Geoffrey Gibney, Michael Atkins. Advances in immunotherapy for melanoma, BMC Medicine, 2016, pp. 20, 14, DOI: 10.1186/s12916-016-0571-0