Progress in Clinical Use of CC Chemokine Receptor 4 Antibody for Regulatory T Cell Suppression


Target antigens

Antibody medicines

Tumor types

Mechanisms of tumor killing

Hematopoietic cell differentiation antigens

CD20

Rituximab

Non-Hodgkin’s lymphoma

ADCC
 
Tositumomab

Non-Hodgkin’s lymphoma

131I
 
Ibritumomab

90Y

CD22

Inotuzumab

Non-Hodgkin’s lymphoma

Ozogamicin

CD30

Brentuximab

Hodgkin’s lymphoma

Monomethyl auristatin E

CD33

Gemtuzumab

Acute myelogenous leukemia

Ozogamicin

CD52

Alemtuzumab

Chronic lymphocytic leukemia

CDC

Chemokine receptor

CCR4

Mogamulizumab

Adult T cell leukemia/lymphoma

ADCC

Tyrosine kinase receptors

EGFR

Cetuximab, panitumumab, nimotuzumab, and 806

Glioma, lung, breast, colon, and head and neck tumors

Inhibition of growth signaling, ADCC and CDC

ERBB2 (HER2)

Trastuzumab and pertuzumab

Breast, colon, lung, ovarian, and prostate cancers

ADCC

ERBB3

MM-121

Breast, colon, lung, ovarian, and prostate cancers

Inhibition of growth signaling

VEGFR

IM-2C6 and CDP791

Epithelium-derived solid tumors

Inhibition of growth signaling

MET

AMG 102 and METMAB

Breast, ovary and lung cancers

Inhibition of growth signaling

IGF1R

AVE1642, IMC-A12, MK-0646, and R1507

Glioma, lung, breast, head and neck, prostate, and thyroid cancers

Inhibition of growth signaling

EPHA3

IIIA4

Lung, kidney and colon cancers, melanoma and glioma

Disrupting the tumor stroma and microvasculature

Death receptors

TRAILR1 (DR3)

Mapatumumab

Colon, lung and pancreas tumors and hematological malignancies

Death signal

TRAILR2 (DR4)

HGS-ETR2 and CS-1008

RANKL

Denosumab

Prostate cancer and bone metastases

Blockade of RANKL/RANK interaction

Glycoproteins

CEA

Labetuzumab

Breast, colon, and lung cancers

131I

EpCAM

Adecatumumab

Breast and prostate cancers

ADCC and CDC

CA-125

Oregovomab

Ovarian cancer

Enhancement of tumor-specific humoral and cellular immune responses

CAIX

cG250

Renal cell carcinoma

131I, 90Y

Others

VEGF

Bevacizumab

Tumor vasculature

Disrupting the tumor microvasculature

Integrin aVb3

Etaracizumab

Tumor vasculature

Integrin a5b1

Volociximab

Tumor vasculature

FAP

Sibrotuzumab

Colon, breast, lung, pancreas, and head and neck cancers

Disrupting the tumor stromal fibroblasts


ADCC antibody-dependent cellular cytotoxicity, CAIX carbonic anhydrase IX, CCR4 CC chemokine receptor 4, CDC complement-dependent cytotoxicity, CEA carcinoembryonic antigen, EGFR epidermal growth factor receptor, EpCAM epithelial cell adhesion molecule, EPHA3 ephrin receptor A3, FAP fibroblast activation protein, HER2 human epidermal growth factor receptor 2, IGF1R insulin-like growth factor 1 receptor, RANK receptor activator of nuclear factor-κB, RANKL RANK ligand, TRAILR tumor necrosis factor-related apoptosis-inducing ligand receptor, VEGF vascular endothelial growth factor, VEGFR VEGF receptor



In contrast, target antigens recognized by antibodies listed in Table 17.2 are immunomodulatory receptors or ligands (Pardoll 2012; Weber 2010; Ngiow et al. 2011a). These receptors, such as cytotoxic T lymphocyte-associated antigen 4 (CTLA-4; also known as CD152), programmed cell death protein 1 (PD-1; also known as CD274), and lymphocyte activation gene 3 (LAG3), are expressed in activated T cells (Schwartz 1992; Lenschow et al. 1996; Rudd et al. 2009; Ishida et al. 1992), and the ligands, including CD80 (also known as B7-1), CD86 (also known as B7-2), and PD-1 ligand 1 (PD-L1, also known as B7-H1 and CD273), are commonly expressed on antigen-presenting cells (APC), tumors, or non-transformed cells in the tumor microenvironment (Zou and Chen 2008). Over-expression of these molecules and their receptor/ligand pathways are a major cause of exhaustion in CD8 T cells. Exhausted CD8 T cells are functionally deficient and have decreased proliferative capacity, cytokine production, and cytotoxic activity and are metabolically deficient (Barber et al. 2006). The antibodies can block the receptor/ligand pathway between infiltrating T cells in tumors and the tumor cells to restore T cell cytotoxic activity against the tumors. Indeed, significant clinical effects by single drug therapy has been reported for ipilimumab (anti-CTLA-4), nivolumab/BMS‑936558 (anti-PD-1), lambrolizumab/MK-3475 (anti-PD-1), and BMS-936559 (anti-PD-L1) (Hodi et al. 2010; Topalian et al. 2012; Hamid et al. 2013; Brahmer et al. 2012). Thus, turning the immune balance to an active state by blocking immune inhibitory signals with antagonistic antibodies is a new direction for the development of antibody drug therapy against tumors.


Table 17.2
Therapeutic antibodies targeting immune checkpoints














































































Antibodies

Targets

Counterparts

Antigens

Cells

Antigens

Cells

Ipilimumab

CTLA-4

T cell

CD80 or CD86 (B7-1) (B7-2)

APC

Tremelimumab

Nivolumab (BMS-936558)

PD-1

T-cell

PD-L1 or PD-L2 (B7-H1) (B7-DC)

APC

Pembrolizumab (MK-3475)

CT-011

AMP-244

BMS-936559

PD-L1 (B7-H1)

Tumor

PD-1, CD80

T cell

MEDI4736

T cell

BMS-986016

LAG3

T cell

MHCII

APC

MGA271

B7-H3

APC

?

T cell ?

Under development

B7-H4

APC

?

T cell ?

Under development

TIM3

T cell

Galectin 9

Tumor

BMS-986015

KIR

T cell

MHCI

Tumor


APC antigen-presenting cell, CTLA-4 cytotoxic T lymphocyte-associated antigen, LAG3 lymphocyte activation gene 3, MHCI major histocompatibility complex class I, MHCII major histocompatibility complex class II, PD-1 programmed cell death protein 1, PD-L PD-1 ligand, TIM3 T cell membrane protein 3, ? unknown

Regulation of regulatory T cells (Tregs) is also considered important for the development of new therapeutics against tumors because many reports indicate infiltrating Tregs in tumors suppress tumor immune responses and Treg depletion by administration of CD25 mAb causes tumor regression in a mouse tumor model (Onizuka et al. 1999; Ohmura et al. 2008). CCR4 is commonly expressed on Tregs; thus, mogamulizumab can diminish not only ATLL cells but also normal Tregs in vivo (Yano et al. 2007; Ishida and Ueda 2006, 2011; Ishida et al. 2003). Therefore, mogamulizumab might be a unique regulator of immune balance to resolve tumor immune suppression, resulting in new therapeutics for tumors. This review focuses on the regulation of Tregs by mogamulizumab and discusses the theory and possibility of a new tumor therapy using mogamulizumab compared with antibodies to immune checkpoint-related molecules such as ipilimumab, nivolumab, and BMS-936559.



17.2 Regulation of Immune Balance in Immune Checkpoints


Immune checkpoints during the sequential steps of immune responses are crucial events to maintain self-tolerance and to protect tissues from damage caused by hyperimmune reactions during pathogenic inflammation (Pardoll 2012; Zou and Chen 2008). CTLA-4 is transiently expressed on effector T cells after activation and functions as an inhibitory receptor in the first immune checkpoint, which regulates antigen presentation from dendritic cells (DC) in primary or secondary lymph nodes (Zou and Chen 2008). When antigen is presented by DCs to naïve or memory T cells, T cells recognize antigen by binding to the T cell receptor (TCR) and epitope peptide complexed with major histocompatibility complex (MHC) expressed on the DC. Simultaneously, co-stimulatory receptor CD28, exclusively expressed on T cells, binds to CD80/CD86 on DCs and stimulates immune responses (Zou and Chen 2008). CTLA-4 shares identical ligands, CD80/CD86, with CD28, and counteracts the activity of CD28 (Schwartz 1992; Rudd et al. 2009). Although the mechanisms of T cell suppression by CTLA-4 are not clear, two different mechanisms have been demonstrated. CTLA-4 is thought to compete with CD28 in binding CD80 and/or CD86 because the binding affinity of CTLA-4 for both ligands is higher than that of CD28 (Linsley et al. 1994; Egen and Allison 2002). Another explanation is the activation of protein phosphatases, Src homolog domain-containing phosphatase 2 (SHP2, also known as PTPN11) and protein phosphatase 2 (PP2, also known as PP2A), which inhibit kinase signaling downstream of TCR and CD28 (Schwartz 1992). CTLA-4 is constitutively expressed on Tregs as well as CD8+ cytotoxic T cells and CD4+ T helper (Th) cells, and is a target gene of the forkhead transcription factor FOXP3 (Hori et al. 2003; Fontenot et al. 2003), the master gene of Treg. FOXP3 regulates the differentiation and functions of Treg (Hill et al. 2007; Gavin et al. 2007) and is one of the mechanisms of effector T cell suppression that downregulates CD80 and CD86 on APCs. This is caused by the binding of CTLA-4 on Treg counteracting CD28 binding CD80/CD86, resulting in the inhibition of antigen presentation to T cells (Vignali et al. 2008) (Fig. 17.1).

A307883_1_En_17_Fig1_HTML.gif


Fig. 17.1
T cell exhaustion mechanisms induced by regulatory T cells (Tregs). Tregs constitutively express cytotoxic T lymphocyte-associated antigen (CTLA)4. The affinity of CTLA-4 to CD80 or CD86 is higher than CD28, thus CTLA-4 outcompetes CD28, and co-stimulatory signals via CD28 are blocked in antigen-stimulated T cells, resulting in anergy. Tregs highly express programmed cell death protein 1 (PD-1), the proliferation of Tregs is enhanced in the presence of ligand, and the effector T cells are exhausted by the release of interleukin (IL)-10, IL-35, and transforming growth factor (TGF)-β from Tregs

PD-1 is another important co-inhibitory receptor stably expressed on effector-phase T cells, which regulates T cell activation by interactions with PD-L1 and/or PD-L2 expressed on tumor cells, APC, and non-transformed cells in the tumor microenvironment (Dong et al. 2002; Konishi et al. 2004; Curiel et al. 2003; Kuang et al. 2009). PD-1 engagement inhibits kinase signaling downstream of TCR through phosphatase SHP2 (Dong et al. 2002). PD-1 is also highly expressed on Tregs and enhances the proliferation of Tregs in the presence of ligand (Freeman et al. 2000; Francisco et al. 2009).

As well as the molecules described above, various inhibitory receptors and ligands are involved in immune checkpoints. T cell membrane protein 3 (TIM3), LAG3 (also known as CD233), B and T Lymphocyte attenuator (BTLA; also known as CD272), adenosine A2a receptor (A2aR), and the family of killer inhibitory receptors are inhibitory receptors expressed on T cells. Galectin 9, MHC class II, herpesvirus entry mediator (HVEM), adenosine, and subsets of human leukocyte antigens (HLA) have been respectively identified as their ligands (Pardoll 2012). These receptors are also expressed on activated T cells, and the ligands are expressed on many types of tumor cells (Ngiow et al. 2011a; Grosso et al. 2007; Cedeno-Laurent and Dimitroff 2012; Fourcade et al. 2012; Baixeras et al. 1992). Furthermore, antibodies to these enhance anti-tumor immunity in mouse models and human in vitro experiments (Baghdadi et al. 2013; Ngiow et al. 2011b). LAG3 is expressed on Tregs as well as effector T cells and enhances Treg functions. A2aR functions in the development of Tregs by inducing FOXP3 expression in CD4+ T cells (Huang et al. 2004; Zarek et al. 2008).


17.3 Clinical Application of the Blockade of Immune Checkpoints


Thus, co-inhibitory receptors/ligands regulate immune responses of effector T cells at sites of inflammation or tumors (Fig. 17.2), and drugs, such as antibodies and small molecules, which block receptor/ligand interactions are expected to be novel beneficial drugs for tumor therapy (Fig. 17.3). Clinical trials of anti-CTLA-4 humanized mAb, ipilimumab, and tremelimumab were conducted from 2000. Although objective clinical responses were observed in ~10 % of patients with melanoma using both antibodies, no survival benefit was observed in comparison with standard melanoma chemotherapy treatment (dacarbazine) in a randomized phase III trial for tremelimumab (Ribas 2010). However, in a randomized, three-arm phase III trial of patients with advanced melanoma using (1) gp100 peptide vaccine alone, (2) gp100 plus ipilimumab, or (3) ipilimumab alone, 3.5-month survival benefit was observed in the ipilimumab alone group compared with the gp100 peptide vaccine alone or gp100 plus ipilimumab groups (Hodi et al. 2010). Furthermore, Grade 3 or 4 immune-related adverse events (AEs) occurred in 10–15 % of patients treated with ipilimumab, including deaths associated with immune-related AEs. Ipilimumab was the first drug to demonstrate a survival benefit for metastatic melanoma, and was approved by the US Food and Drug Administration (FDA) for the treatment of advanced melanoma in 2010. Both humanized mAb to PD-1 (nivolumab/BS936558) and PD-L1 (BS936559) demonstrated very promising results in a phase I clinical trial for multiple tumors (Topalian et al. 2012; Brahmer et al. 2012). Objective response rates to BMS936558 were observed in 18 % of patients with non-small cell lung cancer (14 of 76 patients), 28 % of patients with melanoma (26 of 94 patients), and 27 % of patients with renal cell cancer (9 of 33 patients). Interestingly, no objective response was observed in 17 patients with PD-L1-negative tumors, although 9 of 25 patients (36 %) with PD-L1-positive tumors had an objective response. In a phase I trial using anti-PD-L1 (BMS936559), an objective response (a complete or partial response) was observed in 17 % of patients with melanoma (9 of 52), 12 % of patients with renal cell cancer (2 of 17), 10 % of patients with non-small cell lung cancer (5 of 49), and 6 % of patients with ovarian cancer (1 of 17). Furthermore, responses were durable for both antibodies. Responses lasting 1 year or more were shown in more than half of the patients with 1 year or more of follow-up. Immune-related AEs occurred in 14 % of patients treated with anti-PD-1 and 9 % of patients treated with anti-PD-L1. Interestingly, immune-related AEs frequently occurred in the skin and intestine in patients receiving any of ipilimumab, nivolumab, or BMS936559.

A307883_1_En_17_Fig2_HTML.gif


Fig. 17.2
Immune checkpoint determines the balance of stimulatory and inhibitory signals and the future state of T cells. After T cells recognize antigen epitope peptides presented on human leukocyte antigen (HLA) class I molecules, the signal is transduced in addition to co-stimulatory and co-inhibitory molecule signaling. Co-stimulatory receptors (CD28, CD137, ICOS, OX40, CD27) and co-inhibitory receptors [cytotoxic T lymphocyte-associated antigen (CTLA)-4, programmed cell death protein 1 (PD-1), T cell membrane protein 3 (TIM3), killer cell immunoglobulin-like receptor (KIR), lymphocyte activation gene 3 (LAG3), adenosine A2a receptor (A2aR)] are expressed on T cells, and their co-stimulatory ligands (CD80, CD86, CD137L, B7PR1, OX40L, CD70) and co-inhibitory ligands [CD80, CD86, PD-1 ligand (PD-L) 1, PD-L2, galectin 9, major histocompatibility complex (MHC) class II, adenosine) are expressed on antigen-presenting cells or tumor cells. The balance of stimulatory and inhibitory signals determines whether T cells are activated or exhausted. Once tumor-specific T cells are activated, they elicit killing activity to the tumor cells


A307883_1_En_17_Fig3_HTML.gif


Fig. 17.3
Antibodies to immune checkpoint molecules block co-inhibitory signals turning the immune balance to an active state. In the tumor site, the immune balance is considered inhibitory, caused by inhibitory signals that exceed stimulatory signals. Antagonistic antibodies such as ipilimumab [anti-cytotoxic T lymphocyte-associated antigen (CTLA)-4], nivolumab [anti-programmed cell death protein 1 (PD-1)], lambrolizumab (anti-PD-1), and BMS936559 [anti-PD-1 ligand 1 (PD-L1)] block co-inhibitory signals of T cells, turning the immune balance towards an active state. Exhausted T cells in the tumor site show restored tumor killing functions and kill tumor cells


17.4 Immune Tolerance in Tumors Caused by Regulatory T Cells


It is clarified that FOXP3 elicits Treg functions (Hori et al. 2003; Fontenot et al. 2003). Because FOXP3 knockout mice develop autoimmune diseases involving multiple organs, and patients with FOXP3 homozygous mutations develop immunodysregulation, polyendocrinopathy, enteropathy, and X-linked (IPEX) syndrome (Fontenot et al. 2003; Bennett et al. 2001), Tregs are considered to be crucial for the maintenance of self-tolerance and for maintaining immune balance to prevent damage of healthy cells from excessive immune reactions in cooperation with the immune checkpoint system described above. It was reported that Tregs are recruited and accumulate in tumor tissues by chemokine-induced chemotaxis via interaction between CCR4 and its ligands produced by cells of the tumor microenvironment (Curiel et al. 2004; Zou 2006). Hodgkin’s lymphoma (HL) is a representative example of Treg recruitment to the tumor site by chemokine-induced chemotaxis resulting in immunosuppression. HL is characterized by the presence of a small number of tumor cells in a rich background of T and B cells, macrophages, and other inflammatory cells (Küppers 2009). The question of why a very small number of HL cells can survive in the presence of a large excess of host immune cells was unclear. HL cells express high levels of the CCR4 ligand and thymus and activation-regulated chemokine (TARC)⁄CCL17 (van den Berg et al. 1999), and elevated serum levels of TARC⁄CCL17 were reported as an unfavorable prognostic factor in patients with HL (Weihrauch et al. 2005). Considering these findings, we have shown that HL tumor cells attracted CD4+CCR4+ T cells by interactions with TARC⁄CCL17 and macrophage-derived chemokine (MDC)⁄CCL22. Migratory CD4+ cells attracted by HL tumor cells were hyporesponsive to TCR stimulation and suppressed the activation and proliferation of effector CD4+ T cells in an autologous setting in vitro. Furthermore, double staining showed that HL cells in the affected lymph nodes were surrounded by a large number of lymphocytes expressing both CCR4 and FOXP3 (Ishida et al. 2006). Collectively, these findings imply that the migratory cells induced by HL function as Tregs to create a favorable environment for the tumor cells to escape from host immunity. It was proposed that Treg-mediated immunosuppression is a crucial tumor immune-evasion mechanism in ovarian, gastric, breast, and pancreatic cancers, and may be one of the main obstacles to successful tumor immunotherapy (Curiel et al. 2004; Zou 2006). Therefore, Tregs are a potential target for novel tumor therapy by inhibition of immunosuppression in the tumor site.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Dec 10, 2016 | Posted by in ONCOLOGY | Comments Off on Progress in Clinical Use of CC Chemokine Receptor 4 Antibody for Regulatory T Cell Suppression

Full access? Get Clinical Tree

Get Clinical Tree app for offline access