1
|
Liu Y, Zheng P. Preserving the CTLA-4 Checkpoint for Safer and More Effective Cancer Immunotherapy. Trends Pharmacol Sci 2019; 41:4-12. [PMID: 31836191 PMCID: PMC7210725 DOI: 10.1016/j.tips.2019.11.003] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/08/2019] [Accepted: 11/11/2019] [Indexed: 01/07/2023]
Abstract
A major paradigm in cancer immunotherapy is the use of checkpoint inhibitors to break regulatory mechanisms that usually guard the host against autoimmune diseases. CTLA-4-targeting immunotherapy was the first example that helped establish this paradigm. However, the clinically tested anti-CTLA-4 antibodies exhibit suboptimal efficacy but high toxicity. Recent studies have demonstrated that immunotherapy-related adverse events (irAE) and the cancer immunotherapeutic effect (CITE) represent distinct and therapeutically separable activities of anti-CTLA-4 antibodies. The former is attributable to inactivation of the CTLA-4 checkpoint, while the latter is due to selective depletion of regulatory T cells (Treg) in a tumor microenvironment. Here we argue that for safer and more effective CTLA-4-targeting immune therapy, one should preserve rather than inhibit the CTLA-4 checkpoint while enhancing the efficacy and selectivity of Treg depletion in a tumor microenvironment.
Collapse
Affiliation(s)
- Yang Liu
- Division of Immunotherapy, Institute of Human Virology and Department of Surgery, University of Maryland, Baltimore, MD 21201, USA; OncoImmune, Inc., Rockville, MD 20850, USA.
| | - Pan Zheng
- Division of Immunotherapy, Institute of Human Virology and Department of Surgery, University of Maryland, Baltimore, MD 21201, USA; OncoImmune, Inc., Rockville, MD 20850, USA.
| |
Collapse
|
2
|
Philipp Bewersdorf J, Stahl M, Zeidan AM. Immune checkpoint-based therapy in myeloid malignancies: a promise yet to be fulfilled. Expert Rev Anticancer Ther 2019; 19:393-404. [PMID: 30887841 PMCID: PMC6527485 DOI: 10.1080/14737140.2019.1589374] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 02/27/2019] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Immune system evasion is essential for tumor cell survival and is mediated by the immunosuppressive tumor microenvironment and the activation of inhibitory immune checkpoints. While immune checkpoint-based therapy yielded impressive results in several advanced solid malignancies such as melanoma and non-small cell lung cancer, its role in acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) is still evolving. Areas covered: Here we review the immunology in the tumor microenvironment in the bone marrow and discuss the current preclinical and clinical data for immune checkpoint-based therapy in myeloid neoplasms. Expert commentary: Clinical trials of immune checkpoint inhibitors (ICI) in AML and MDS are still in early stages and reported results so far have been modest especially for monotherapy use in the refractory settings. However, there are preliminary data for synergistic effects for combination of multiple ICI with hypomethylating agents and conventional chemotherapy. ICI might also be effective in eradicating minimal residual disease and to prevent relapse following induction chemotherapy or hematopoietic stem cell transplant. Additional trials to provide insight into the efficacy and safety profile of immune checkpoint-based therapy, its optimal timing and potential combination with other types of therapy as well as identification of predictive biomarkers are needed.
Collapse
Affiliation(s)
- Jan Philipp Bewersdorf
- Department of Internal Medicine, Section of Hematology, Yale University School of Medicine, New Haven, CT
| | - Maximilian Stahl
- Department of Medicine, Section of Hematologic Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Amer M Zeidan
- Department of Internal Medicine, Section of Hematology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
3
|
Chang Z, Song R, Xu S, Xu M, Yu X. CD 152 gene polymorphisms and risk of osteosarcoma in Chinese population. Tumour Biol 2014; 35:6809-14. [PMID: 24729087 DOI: 10.1007/s13277-014-1891-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 03/25/2014] [Indexed: 10/25/2022] Open
Abstract
Osteosarcoma has become a health threat for adolescents and young adults. To identify the genetic risk factor for the malignancy is in urgent need. Several studies have investigated the role of CD 152 polymorphisms in osteosarcoma in a sample of Chinese population. However, the association is poorly defined due to lack of a sufficiently large sample. In this study, we performed a meta-analysis of all CD 152 polymorphisms that had been implicated in osteosarcoma to examine the association. We searched the electronic MEDLINE database until December 31, 2013, to identify the studies regarding the association between CD 152 polymorphisms and osteosarcoma. Inclusion criteria were followed in the selection of eligible study. The genotypic and allelic data were collected from all studies included to evaluate the risk of osteosarcoma (odds ratio, OR). We found statistically significant evidence of the studied CD 152 polymorphisms and increased risk of osteosarcoma in homozygous (OR = 1.79, 95 % CI = 1.40-2.29, P = 0.958), recessive (OR = 1.77, 95 % CI = 1.40-2.25, P = 0.899), and allele model (OR = 1.21, 95 % CI = 1.09-1.34, P = 1.000). This increased risk was also revealed in single nucleotide polymorphism (SNP) +49G>A and SNP 326G>A. Our meta-analysis indicates that there may be an association between CD 152 polymorphisms and risk of osteosarcoma in Chinese population. Further validation of the observation is necessary.
Collapse
Affiliation(s)
- Zhengqi Chang
- Department of Orthopedics, General Hospital of Jinan Military Commanding Region, Jinan, 250031, Shandong Province, China
| | | | | | | | | |
Collapse
|
4
|
Houshmand B, Rafiei A, Hajilooi M. Influence of cytotoxic T lymphocyte antigen-4 (CTLA-4) gene polymorphisms in periodontitis. Arch Oral Biol 2012; 57:1218-24. [DOI: 10.1016/j.archoralbio.2012.03.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Revised: 03/04/2012] [Accepted: 03/11/2012] [Indexed: 10/28/2022]
|
5
|
Kämmerer PW, Toyoshima T, Schöder F, Kämmerer P, Kuhr K, Brieger J, Al-Nawas B. Association of T-cell regulatory gene polymorphisms with oral squamous cell carcinoma. Oral Oncol 2011; 46:543-8. [PMID: 20435510 DOI: 10.1016/j.oraloncology.2010.03.025] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Revised: 03/30/2010] [Accepted: 03/30/2010] [Indexed: 10/19/2022]
Abstract
Costimulatory molecules have complementary effects on T-cell activation and their balance may control the development of oral cancer. The aim of this study was to determine the relevance of cytotoxic T-lymphocyte antigen 4 (CTLA-4), CD28 and inducible costimulator (ICOS) polymorphisms in oral squamous cell carcinoma (OSCC). Genotyping for CTLA-4 (-1661 A/G and +49 A/G), CD28 (0 C/G and +3160 G/T) and ICOS (+637 A/C and +1599 C/T) was performed in the 83 patients with OSCC, compared to the 40 unrelated healthy volunteers as controls. The genotype CTLA-4 -1661 was significantly different between the patient group and the control group. The allele CTLA-4 -1661 G was significantly found more frequent in patients with OSCC (p=0.001). In bivariate analysis, noticeable differences between OSCC and controls were seen. The combinations CTLA-4 -1661 G/G and CTLA-4 +49 A/G, ICOS +1559 C/T and ICOS +1559 C/C each with CTLA-4 -1661 G/G, ICOS +637 C/C and ICOS +637 A/C each with CTLA-4 -1661, CTLA-4 -1661 A/G and ICOS +637 C/C, CD28 +3160 G/T and CTLA-4 -1661 A/A and CD28 +3160 G/T and CTLA-4 -1661 A/G were seen in the patient group only. Especially the polymorphisms of the CTLA-4 -1661-genotype - alone and in combination with other T cell regulator polymorphisms - seem to be possible predisposing factors for OSCC. Therefore, they might be future targets for a primary prophylaxis or an individualised therapy.
Collapse
Affiliation(s)
- Peer W Kämmerer
- Department of Oral and Maxillofacial Surgery, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | | | | | | | | | | | | |
Collapse
|
6
|
Wang L, Liu JQ, Talebian F, El-Omrani HY, Khattabi M, Yu L, Bai XF. Tumor expression of CD200 inhibits IL-10 production by tumor-associated myeloid cells and prevents tumor immune evasion of CTL therapy. Eur J Immunol 2010; 40:2569-79. [PMID: 20662098 DOI: 10.1002/eji.201040472] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
CD200 is a cell-surface glycoprotein that functions through interaction with the CD200 receptor on myeloid lineage cells to regulate myeloid cell functions. Expression of CD200 has been implicated in multiple types of human cancer; however, the impact of tumor expression of CD200 on tumor immunity remains poorly understood. To evaluate this issue, we generated CD200-positive mouse plasmacytoma J558 and mastocytoma P815 cells. We found that established CD200-positive tumors were often completely rejected by adoptively transferred CTL without tumor recurrence; in contrast, CD200-negative tumors were initially rejected by adoptively transferred CTL but the majority of tumors recurred. Tumor expression of CD200 significantly inhibited suppressive activity and IL-10 production by tumor-associated myeloid cells (TAMC), and as a result, more CTL accumulated in the tumor and exhibited a greater capacity to produce IFN-gamma in CD200-positive tumors than in CD200-negative tumors. Neutralization of IL-10 significantly inhibited the suppressor activity of TAMC, and IL-10-deficiency allowed TAMC to kill cancer cells and their antigenic variants, which prevented tumor recurrence during CTL therapy. Thus, tumor expression of CD200 prevents tumor recurrence via inhibiting IL-10 production by TAMC.
Collapse
Affiliation(s)
- Lixin Wang
- Department of Pathology and Comprehensive Cancer Center, The Ohio State University Medical Center, Columbus, OH 43210, USA
| | | | | | | | | | | | | |
Collapse
|
7
|
Moreira G, Fulgêncio LB, DE Mendonça EF, Leles CR, Batista AC, DA Silva TA. T regulatory cell markers in oral squamous cell carcinoma: Relationship with survival and tumor aggressiveness. Oncol Lett 2010; 1:127-132. [PMID: 22966269 DOI: 10.3892/ol_00000023] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2009] [Accepted: 08/04/2009] [Indexed: 11/06/2022] Open
Abstract
Tumor-infiltrating lymphocytes (TILs) are a heterogeneous cell family which plays an important role in tumor-associated immune response. Of these, T regulatory (Treg) cells have also been shown to inhibit anti-tumor response. We aimed to evaluate the expression of T regulatory cell markers (CD4, CD25, CTLA-4 and FoxP3) in samples of oral cavity squamous cell carcinoma (OCSCC) and lip SCC (LSCC) by immunohistochemistry. The relationship of Treg markers with survival data and the proliferative index were also evaluated. We observed similar numbers of CD4-, CD25- and FoxP3(+) cells in OCSCC and LSCC. On the other hand, numbers of CTLA-4(+) cells were significantly lower in OCSCC than in LSCC. OCSCC samples with high numbers of CD4 exhibited a high proliferative index, while samples with high CTLA-4 counts demonstrated a low tumoral proliferative index. A log-rank test showed that patients with OCSCC that presented high counts of CD4 showed a significantly decreased survival compared with patients with low cell counts. In contrast, high CD25(+) cell counts were associated with increased survival. Our results suggest an association of CD4 with poor prognosis, while CD25 expression is related with favorable prognosis. These findings result from the heterogeneity of TIL subsets that display an antagonistic role in tumor immune cell response.
Collapse
Affiliation(s)
- Geane Moreira
- Department of Oral Surgery and Pathology, Dental School, Federal University of Minas Gerais, Belo Horizonte
| | | | | | | | | | | |
Collapse
|
8
|
Mir MA, Agrewala JN. Signaling through CD80: an approach for treating lymphomas. Expert Opin Ther Targets 2008; 12:969-79. [DOI: 10.1517/14728222.12.8.969] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
9
|
Bai XF, Liu JQ, Joshi PS, Wang L, Yin L, Labanowska J, Heerema N, Zheng P, Liu Y. Different lineages of P1A-expressing cancer cells use divergent modes of immune evasion for T-cell adoptive therapy. Cancer Res 2007; 66:8241-9. [PMID: 16912204 DOI: 10.1158/0008-5472.can-06-0279] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Tumor evasion of T-cell immunity remains a significant obstacle to adoptive T-cell therapy. It is unknown whether the mode of immune evasion is dictated by the cancer cells or by the tumor antigens. Taking advantage of the fact that multiple lineages of tumor cells share the tumor antigen P1A, we adoptively transferred transgenic T cells specific for P1A (P1CTL) into mice with established P1A-expressing tumors, including mastocytoma P815, plasmocytoma J558, and fibrosarcoma Meth A. Although P1CTL conferred partial protection, tumors recurred in almost all mice. Analysis of the status of the tumor antigen revealed that all J558 tumors underwent antigenic drift whereas all P815 tumors experienced antigenic loss. Interestingly, although Meth A cells are capable of both antigenic loss and antigenic drift, the majority of recurrent Meth A tumors retained P1A antigen. The ability of Meth A to induce apoptosis of P1CTL in vivo alleviated the need for antigenic drift and antigenic loss. Our data showed that, in spite of their shared tumor antigen, different lineages of cancer cells use different mechanisms to evade T-cell therapy.
Collapse
Affiliation(s)
- Xue-Feng Bai
- Department of Pathology and Comprehensive Cancer Center, The Ohio State University Medical Center, Columbus, OH 43210, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Chen W, Wang J, Shao C, Liu S, Yu Y, Wang Q, Cao X. Efficient induction of antitumor T cell immunity by exosomes derived from heat-shocked lymphoma cells. Eur J Immunol 2006; 36:1598-607. [PMID: 16708399 DOI: 10.1002/eji.200535501] [Citation(s) in RCA: 147] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Exosomes secreted by tumor cells could serve as a promising immunotherapeutic tumor vaccine. Heat shock proteins (HSP) induced in tumor cells by heat shock are molecular chaperones with potent adjuvant activity in the induction of antigen-specific T cell responses. To improve exosome-based tumor vaccines, we have investigated the efficacy of exosomes derived from heat-shocked mouse B lymphoma cells (HS-Exo) in the induction of antitumor immune responses. We found that HS-Exo, compared with control exosomes derived from the same cells (Exo), contain more HSP60 and HSP90 and increased amounts of molecules involved in immunogenicity including MHC class I, MHC class II, CD40, CD86, RANTES and IL-1beta. Furthermore, HS-Exo induce both phenotypic and functional maturation of dendritic cells more efficiently. HS-Exo immunization activates T cell responses more potently. Importantly, HS-Exo induce dramatically increased antitumor immune responses compared to control exosomes from the same cells in prophylaxis and therapeutic in vivo lymphoma models. We further demonstrate that CD8(+) T cells are the predominant T cell subset responsible for the antitumor effect of HS-Exo and that CD4(+) T cells are necessary in the induction phase of tumor rejection in a prophylaxis model. These findings provide a novel strategy to improve the efficacy of exosome-based tumor vaccines.
Collapse
Affiliation(s)
- Weilin Chen
- The Institute of Immunology, Zhejiang University, Hangzhou, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
11
|
Li O, Chang X, Zhang H, Kocak E, Ding C, Zheng P, Liu Y. Massive and destructive T cell response to homeostatic cue in CD24-deficient lymphopenic hosts. ACTA ACUST UNITED AC 2006; 203:1713-20. [PMID: 16769998 PMCID: PMC2118348 DOI: 10.1084/jem.20052293] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
In response to a lymphopenic cue, T lymphocytes undergo a slow-paced homeostatic proliferation in an attempt to restore T cell cellularity. The molecular interaction that maintains the pace of homeostatic proliferation is unknown. In this study, we report that in lymphopenic CD24-deficient mice, T cells launch a massive proliferation that results in the rapid death of the recipient mice. The dividing T cells have phenotypes similar to those activated by cognate antigens. The rapid homeostatic proliferation is caused by a lack of CD24 on dendritic cells (DCs). Interestingly, although CD24 expression in T cells is required for optimal homeostatic proliferation in the wild-type (WT) host, mice lacking CD24 on all cell types still mount higher homeostatic proliferation than the WT mice. Thus, a lack of CD24 in the non–T host cells bypassed the requirement for T cell expression of CD24 in homeostatic proliferation in the WT host. Our data demonstrate that CD24 expressed on the DCs limits T cell response to homeostatic cue and prevents fatal damage associated with uncontrolled homeostatic proliferation.
Collapse
Affiliation(s)
- Ou Li
- Division of Cancer Immunology, Department of Pathology, The Ohio State University Medical Center, Columbus, OH 43210, USA
| | | | | | | | | | | | | |
Collapse
|
12
|
Tirapu I, Huarte E, Guiducci C, Arina A, Zaratiegui M, Murillo O, Gonzalez A, Berasain C, Berraondo P, Fortes P, Prieto J, Colombo MP, Chen L, Melero I. Low surface expression of B7-1 (CD80) is an immunoescape mechanism of colon carcinoma. Cancer Res 2006; 66:2442-50. [PMID: 16489051 DOI: 10.1158/0008-5472.can-05-1681] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Artificially enforced expression of CD80 (B7-1) and CD86 (B7-2) on tumor cells renders them more immunogenic by triggering the CD28 receptor on T cells. The enigma is that such B7s interact with much higher affinity with CTLA-4 (CD152), an inhibitory receptor expressed by activated T cells. We show that unmutated CD80 is spontaneously expressed at low levels by mouse colon carcinoma cell lines and other transplantable tumor cell lines of various tissue origins. Silencing of CD80 by interfering RNA led to loss of tumorigenicity of CT26 colon carcinoma in immunocompetent mice, but not in immunodeficient Rag-/- mice. CT26 tumor cells bind CTLA-4Ig, but much more faintly with a similar CD28Ig chimeric protein, thus providing an explanation for the dominant inhibitory effects on tumor immunity displayed by CD80 at that expression level. Interestingly, CD80-negative tumor cell lines such as MC38 colon carcinoma and B16 melanoma express CD80 at dim levels during in vivo growth in syngeneic mice. Therefore, low CD80 surface expression seems to give an advantage to cancer cells against the immune system. Our findings are similar with the inhibitory role described for the dim CD80 expression on immature dendritic cells, providing an explanation for the low levels of CD80 expression described in various human malignancies.
Collapse
Affiliation(s)
- Iñigo Tirapu
- Gene Therapy Unit, Department of Medicine, Centro de Investigación Médica Aplicada and Clínica Universitaria, University of Navarra School of Medicine, Pamplona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Wong YK, Chang KW, Cheng CY, Liu CJ. Association of CTLA-4 gene polymorphism with oral squamous cell carcinoma. J Oral Pathol Med 2006; 35:51-4. [PMID: 16393254 DOI: 10.1111/j.1600-0714.2005.00377.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is a worldwide problem. The main mechanism of tumor immunity is the destruction of tumor cells by cytolytic T lymphocytes. Cytotoxic T lymphocyte-associated antigen 4 (CTLA-4; CD152), a negative regulator of T-lymphocyte activation, plays an extremely important role in the immune tolerance and anergy. This study was designed to investigate the role of CTLA-4 polymorphism in OSCC. METHODS The CTLA-4 +49 A/G polymorphism was studied in 118 patients with OSCC and 147 healthy controls by using restriction fragment length polymorphism (RFLP). The genotype and phenotype frequencies were evaluated in Fisher's exact test. RESULTS There was no significant difference in the frequency of CTLA-4 polymorphism between the OSCC study group and healthy controls. The CTLA-4 A/A genotype was significantly associated with a younger age of onset of OSCC (P = 0.04). The AA genotype was associated with significantly poorer survival (P = 0.003). CONCLUSION The present study is the first to shows that the A/A polymorphism is associated with poor survival in OSCC in Taiwan.
Collapse
Affiliation(s)
- Y-K Wong
- Department of Oral and Maxillofacial Surgery, Veterans General Hospital, Taichung, Taiwan
| | | | | | | |
Collapse
|
14
|
Chakrabarti R, Zhou ZF, Chang Y, Prud'homme GJ. A mutant B7-1/Ig fusion protein that selectively binds to CTLA-4 ameliorates anti-tumor DNA vaccination and counters regulatory T cell activity. Vaccine 2005; 23:4553-64. [PMID: 15919138 DOI: 10.1016/j.vaccine.2005.05.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2005] [Accepted: 05/02/2005] [Indexed: 11/27/2022]
Abstract
We have shown that a plasmid encoding a B7-1/Ig fusion protein enhanced DNA vaccination against human carcinoembryonic antigen (CEA) more effectively than the plasmid encoding membrane-bound B7-1. However, it was not known if B7-1/Ig acted only by binding CD28 (amplifying a stimulatory signal) or by blocking CTLA-4 on T cells (removing inhibitory signals). Here, we aimed to determine this using a plasmid encoding mutant B7-1/Ig (B7-1wa/Ig), which binds only to CTLA-4 but not to CD28. Our results showed that both the B7-1/Ig and B7-1wa/Ig plasmids, when co-administered with a CEA plasmid, enhanced tumor rejection and the in vitro anti-CEA response. Therefore, B7-1wa/Ig ameliorates DNA vaccination, presumably by binding to CTLA-4. This could result from a number of non-exclusive mechanisms, such as a reduced threshold for T-cell activation, or blockade of CTLA-4/B7-mediated tolerogenic signals in DCs or T cells. We found that, in vitro, a significant fraction of CD3/CD28-activated T cells (in the absence of DCs) expressed CTLA-4 and B7-1. Primed T cells of CTLA-4(+)B7-1(+/-) phenotype acted as regulatory T cells by inhibiting IFNgamma production by re-stimulated CTLA-4(-)B7-1(-) cells, and this was reversed by antibodies against IL-10 or TGF-beta1. Both B7-1wa/Ig and CTLA-4/Ig, which bind to CTLA-4 and B7-1/B7-2 respectively, enhanced IFNgamma production, but not the proliferation or IL-4 release in mixed T-cell populations containing these two cell types. In contrast, CTLA-4(-)B7-1(-) T cells produced IFNgamma which was not affected by B7-1wa/Ig or CTLA-4/Ig. These results suggest that blocking of CTLA-4/B7-1 binding in T cell/T cell interactions blocks negative regulatory signals. This might be the mechanism, at least in part, of the enhancement of anti-tumor immunity by the B7-1wa/Ig and B7-1/Ig plasmids.
Collapse
Affiliation(s)
- Rabindranath Chakrabarti
- Department of Laboratory Medicine and Pathobiology, St. Michael's Hospital and University of Toronto, 30 Bond Street, Toronto, Ontario, Canada M5B 1W8.
| | | | | | | |
Collapse
|
15
|
Scrivener S, Goddard RV, Kaminski ER, Prentice AG. Abnormal T-cell function in B-cell chronic lymphocytic leukaemia. Leuk Lymphoma 2003; 44:383-9. [PMID: 12688308 DOI: 10.1080/1042819021000029993] [Citation(s) in RCA: 124] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
There is increasing evidence of T cell dysfunction in B cell chronic lymphocytic leukaemia (B-CLL) which may contribute to the aetiology and progress of the disease. An absolute CD8+ lymphocytosis correlates with disease progression and low expression of CD4 and CD8 (as found in autoimmune disease) is seen with abnormal expression of other surface molecules. Although the expression of T cell surface activation markers, CD25 and CD152, may be increased on culture in B-CLL serum, response to the common mitogens, PHA and PWM, is reduced. This and the excess of CD8 cells may explain partly the variable cooperation of T cells with B cell production of immunoglobulin in B-CLL. In the context of T cell cross-talk with antigen presenting cells, B-CLL B cells are poor antigen presenters. But the T cells themselves have significant abnormalities of expression of the many antigens and ligands necessary for this process. In particular, they exhibit variable expression of the low affinity and non-specific adhesion molecules LFA-1 and ICAM-1, variable, clonally restricted and skewed expression of the TCR repertoire (implying repeated antigenic stimulation possibly by CLL antigens), reduced CD28 and CD152 expression (implying impairment of ability to start or stop an immune response) and reduced IL2 and CD25 (IL2 R) expression (critical for positive feed-back in maintenance and expansion of the T cell response to antigen presentation). Although the production of IL2 and other cytokines by the T cell in B-CLL may be impaired, production of the anti-apoptotic cytokine IL4 is not and there may be a unique and expanded subset of CD8/CD30 cells capable of releasing IL4. The relationship of this T cell subset to the malignant B cell in vivo is unknown. However, T cells which are CD4+/CD152+/CCR4+ migrate selectively in vitro in response to the chemokine CCL22 (specific for the receptor CCR4) produced by the malignant B cells and are always seen amongst the malignant cells in bone marrow and lymph nodes from B-CLL patients. Other abnormalities of cytokine secretion are described. These findings suggest that the T cell in B-CLL may be unable to start, maintain and complete an immune response to the malignant B cell and other antigens and may be involved directly in sustaining the tumour. However, autologous tumour specific cytotoxicity has been shown in vitro and T cells which recognise tumour-derived heavy chain fragments circulate in vivo. If adoptive immunotherapy of any nature is to succeed in B-CLL, manipulation to optimise these CTL responses is needed to overcome the profound and variable T cell dysfunction in this disease.
Collapse
MESH Headings
- Antibody Formation
- Antigens, CD/physiology
- Antigens, Neoplasm/immunology
- Antigens, Surface/physiology
- Cell Adhesion Molecules/physiology
- Colony-Forming Units Assay
- Cytokines/metabolism
- Cytotoxicity, Immunologic
- Disease Progression
- Humans
- Immunologic Deficiency Syndromes/etiology
- Immunologic Deficiency Syndromes/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/complications
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Lymphocyte Activation/drug effects
- Lymphocyte Cooperation
- Lymphocyte Count
- Mitogens/pharmacology
- Neoplasm Proteins/immunology
- Neoplasm Proteins/physiology
- Receptor-CD3 Complex, Antigen, T-Cell/immunology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
Collapse
Affiliation(s)
- S Scrivener
- Plymouth Post-graduate Medical School, Derriford Combined Laboratories, Derriford Hospital, Plymouth PL6 8DH, UK
| | | | | | | |
Collapse
|