Programmed cell death protein 1, also known as PD-1 and CD279 (cluster of differentiation 279), is a cell surface receptor that plays an important role in downregulating the immune system and promoting self tolerance by suppressing T cell inflammatory activity. PD-1 is an immune checkpoint and guards against autoimmunity through a dual mechanism of promoting apoptosis (programmed cell death) in antigen specific T-cells in lymph nodes while simultaneously reducing apoptosis in regulatory T cells (anti-inflammatory, suppressive T cells).

The PD-1 protein is encoded in humans by the PDCD1 gene.PD-1 is a cell surface receptor that belongs to the immunoglobulin superfamily and is expressed on T cells and pro-B cells. PD-1 binds two ligandsPD-L1 and PD-L2.

A new class of drugs that block PD-1, PD-1 inhibitors, activate the immune system to attack tumors and are therefore used with varying success to treat some types of cancer. PD-1 inhibitors are often used in conjunction with ant-CTLA4 inhibitors. This form of combination therapy and other methods of treatment involving PD-1 inhibitors are being studied for their potential to treat a range of diseases. 

Function

Several lines of evidence suggest that PD-1 and its ligands negatively regulate immune responses. PD-1 knockout mice have been shown to develop lupus-like glomerulonephritis and dilated cardiomyopathy on the C57BL/6 and BALB/c backgrounds, respectively. In vitro, treatment of anti-CD3 stimulated T cells with PD-L1-Ig results in reduced T cell proliferation and IFN-γ secretion. IFN-γ is a key pro-inflammatory cytokines that promotes T cell inflammatory activity. Reduced T cell proliferation also correlated with attenuated IL-2 secretion, and together, these data suggest that PD-1 negatively regulates T cell responses.

Experiments using PD-L1 transfected DCs and PD-1 expressing transgenic (Tg) CD4+ and CD8+ T cells suggest that CD8+ T cells are more susceptible to inhibition by PD-L1, although this could be dependent on the strength of TCR signaling. Consistent with a role in negatively regulating CD8+ T cell responses, using an LCMV viral vector model of chronic infection, one recent study showed that the PD-1-PD-L1 interaction inhibits activation, expansion and acquisition of effector functions of virus specific CD8+ T cells, which can be reversed by blocking the PD-1-PD-L1 interaction.

Given findings that CTLA-4 negatively regulates anti-tumor immune responses, the closely related molecule PD-1 has been independently explored as a target for immunotherapy.

In one study, researchers analyzed the ability of 2C T cells to recognize and lyse cancerous cells in a B16-F10 melanoma that had been modified to express the antigenic peptide SIYRYYGL. 2C CD8 T cells incubated with IFN-γ treated B16 targets expressing SIYRYYGL peptide poorly lyse their targets and secrete low levels of IL-2. However, PD-1 knockout 2C T cells have heightened cytolytic capacity and IL-2 secretion, suggesting that PD-1 negatively regulates anti-tumor CD8 T cell responses.

Similarly, P815 mastocytoma, which does not express PD-L1 unless treated with IFN-γ, can be transduced to express PD-L1, resulting in inhibition of in vitro CD8-mediated cytotoxicity and enhanced in vivo tumor growth. In vitro cytotoxicity and in vivo inhibition of growth can be restored by anti-PD-L1 antibodies or by genetic ablation of PD-1

Together, these data suggest that expression of PD-L1 on tumor cells inhibits anti-tumor activity through engagement of PD-1 on effector T cells. Expression of PD-L1 on tumors is correlated with reduced survival in esophageal, pancreatic and other types of cancers, highlighting this pathway as a target for immunotherapy. Said et al. showed that binding of the PD-L1 to the PD-1 receptor expressed on monocytes induces IL-10 production which inhibits CD4 T-cell function.

Cancer

Combination therapy using both anti-PD1 along with anti-CTLA4 therapeutics have emerged as important tumor treatments within the field of checkpoint inhibition.

A combination of PD1 and CTLA4 antibodies has been shown to be more effective than either antibody alone in the treatment of a variety of cancers. The effects of the two antibodies do not appear to be redundant.[1] [2] [3] Anti-CTLA4 treatment leads to an enhanced antigen specific T cell dependent immune reaction while anti-PD-1 appears to reactivate CD8+ T cells ability to lyse cancer cells.[4][5]

In clinical trials, combination therapy has been shown to be effective in reducing tumor size in patients that are unresponsive to single co-inhibitory blockade, despite increasing levels of toxicity due to anti-CTLA4.[6] A recent study has demonstrated that a combination of PD1 and CTLA4 induced up to a ten-fold higher number of CD8+ T cells that are actively infiltrating the tumor tissue.[7] The authors hypothesized that the higher levels of CD8+ T cell infiltration was due to anti-CTLA-4 inhibited the conversion of CD4 T cells to T regulator cells and further reduced T regulatory suppression with anti-PD-1. This combination promoted a more robust inflammatory response to the tumor that reduced the size of the cancer. Most recently, the FDA has approved a combination therapy with both anti-CTLA4 (ipilimumab) and anti-PD1 (nivolumab) in October 2015.[8]

The molecular factors and receptors necessary making a tumor receptive to anti-PD1 treatment remains unknown. PDL1 expression on the surface on cancer cells plays a significant role and a recent study confirmed that PDL1+ tumors were twice as likely to respond to combination treatment.[9] [10] However subsequent investigations have indicated that patients with PDL1- also have limited, responses to anti-PD1, demonstrating that PDL1 expression is not an absolute determinant of the effectiveness of therapy.[11]

Researchers have correlated higher mutational burden in the tumor to a greater effect of the anti-PD-1 treatment. In clinical trials, patients who benefited from anti-PD1 treatment had cancers, such as melanoma, bladder cancer, and gastric cancer, that had a median higher average number of mutations than the patients who do did not respond to the therapy. However, the correlation between higher tumor burden and the clinical effectiveness of PD-1 immune blockade is still uncertain.[12]

HIV Therapy

Drugs targeting PD-1 in combination with other negative immune checkpoint receptors, such as (TIGIT), may augment immune responses and/or facilitate HIV eradication.[13] [14] T lymphocytes exhibit elevated expression of PD-1 in cases of chronic HIV infection. Heightened presence of the PD-1 receptors corresponds to exhaustion of the HIV specific CD8+ cytotoxic and CD4+ helper T cell populations that are vital in combating the virus. Immune blockade of PD-1 resulted in restoration of T cell inflammatory phenotype necessary to combat the progression of disease.[15][16]

Alzheimer's Disease

Blockade of PD-1 has been correlated to a reduction in cerebral amyloid-β plaques and improves cognitive performance in mice. In a recent study, researchers determined that the immune blockade of PD-1 evoked an IFN-γ dependent immune response that recruited monocyte-derived macrophages to the brain, who were capable of clearing the amyloid-β plaques from the tissue. Repeated administrations with anti-PD-1 were found to be necessary to maintain the therapeutic effects of the treatment. The researchers concluded that the amyloid fibrils are in fact immunosuppressive and this finding has been separately confirmed by several other studies by the Steinman group examining the therapeutic effects of the fibrils in neuroinflammatory diseases.[17][18][19] PD-1 counteracts the effects the effects of the fibrils by boosting immune activity and triggering an immune pathway that allows for brain repair.[20] Additionally, impairment of the PD-1 pathway is correlated with the symptoms of Alzheimer's disease.[21]

With my article, I plan to add much more detail to the entry on PD-1 given its recent emergence as one of most central pathways that immunologists and clinicians are targeting as part of checkpoint inhibition treatments of a wide variety of cancers. I have found many recent reviews and articles that cover exciting advancements in the field and expanded understanding of the cellular mechanism of anti-PD-1 treatments within the past few years. I plan to continue collecting reliable reviews and, but here is current list of what I have found and will form the foundation of my edits for the wikipedia article.

Baruch, K., Deczkowska, A., Rosenzweig, N., Tsitsou-Kampeli, A. Sharif. (2016). "PD-1 immune checkpoint blockade reduces pathology and improves memory in mouse models of Alzheimer's disease". Nature Medicine. Vol. 22. No. 2. 135–137.

Chen, Daniel S., and Mellman, Ira. (2013) Oncology meets immunology: the cancer immunity cycle. Cell Immunity. 39.

Curran, Michael A., Montalvo, Welby, Yagita, Hideo, and Allison, James P. (2010) PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. PNAS. Vol. 107 No. 9, 4275-4280.

Herbst, Roy S., Soria, Jean-Charles, Kowanetz, Marcin, Fine, Greg D. (2014) Predictive correlates of response to anti-PD-L1 in cancer patients. Nature. Vol. 0, 1-6.

Kurnellas, M. P., Ghosn, E. E., Schartner, J. M., Baker, J. (2015). Amyloid Fibrils activate B-1a lymphocytes to ameliorate inflammatory brain disease. PNAS. Vol. 112. No. 49.

Restifio, Nicholas P. (2013) A “big data” view of the tumor “immunome.” Cell Immunity. 39.

Topalian, Suzanne, Sharpe, Arlene H. (2014). Balance and imbalance in the immune system: life on the edge. Cell Immunity. 41.

Topalian, Suzanne L., Taube, Janis M., Anders, Robert A., Pardoll, Drew M. (2016). Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nature. Vol. 16, 275-287.

Wang, Jun, Yuan, Ruirong, Song, Wenru, Sun, Jingwei, Liu, Delong, Li, Zihai. (2017) PD-1, PD-L1 (B7-H1) and tumor site immune modulation therapy: the historical perspective. J. Hema. and Onco. 

Zaretsky, Jesse M., Garcia-Diaz, Angel, Shin, Daniel S., Escuin-Ordinas, Helena, Hugo, Willy, Hu-Lieskovan, Siwen. (2016) Mutations associated with acquired resistance to PD-1 blockade in melanoma. New England Journal of Medicine. Vol. 375, No. 9, 819-829.

Zi-Ning Zhang,Ming-Lu Zhu, Yan-Hong Chen,1,2 Ya-Jing Fu. (2015). Elevation of TIM-3 and PD-1 expression on T cells appears early in HIV infection and differential TIM-3 and PD-1 expression patterns can be induced by common γ-cytokines. BioMed Research Inter.  Vol. 2015.

  1. ^ Herbst, Roy S.; Soria, Jean-Charles; Kowanetz, Marcin; Fine, Gregg D.; Hamid, Omid; Gordon, Michael S.; Sosman, Jeffery A.; McDermott, David F.; Powderly, John D. (2014-11-27). "Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients". Nature. 515 (7528): 563–567. doi:10.1038/nature14011. ISSN 1476-4687. PMC 4836193. PMID 25428504.{{cite journal}}: CS1 maint: PMC format (link)
  2. ^ Snyder, Alexandra; Makarov, Vladimir; Merghoub, Taha; Yuan, Jianda; Zaretsky, Jesse M.; Desrichard, Alexis; Walsh, Logan A.; Postow, Michael A.; Wong, Phillip (2014-12-04). "Genetic Basis for Clinical Response to CTLA-4 Blockade in Melanoma". New England Journal of Medicine. 371 (23): 2189–2199. doi:10.1056/NEJMoa1406498. ISSN 0028-4793. PMC 4315319. PMID 25409260.{{cite journal}}: CS1 maint: PMC format (link)
  3. ^ Buchbinder, Elizabeth I.; Desai, Anupam (2016-02-01). "CTLA-4 and PD-1 Pathways: Similarities, Differences, and Implications of Their Inhibition". American Journal of Clinical Oncology. 39 (1): 98–106. doi:10.1097/COC.0000000000000239. ISSN 1537-453X. PMC 4892769. PMID 26558876.{{cite journal}}: CS1 maint: PMC format (link)
  4. ^ Curran, Michael A.; Montalvo, Welby; Yagita, Hideo; Allison, James P. (2010-03-02). "PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors". Proceedings of the National Academy of Sciences. 107 (9): 4275–4280. doi:10.1073/pnas.0915174107. ISSN 0027-8424. PMC 2840093. PMID 20160101.{{cite journal}}: CS1 maint: PMC format (link)
  5. ^ Sliwkowski, Mark X.; Mellman, Ira (2013-09-13). "Antibody Therapeutics in Cancer". Science. 341 (6151): 1192–1198. doi:10.1126/science.1241145. ISSN 0036-8075. PMID 24031011.
  6. ^ Chen, Daniel S.; Mellman, Ira (2013-07-25). "Oncology meets immunology: the cancer-immunity cycle". Immunity. 39 (1): 1–10. doi:10.1016/j.immuni.2013.07.012. ISSN 1097-4180. PMID 23890059.
  7. ^ Curran, Michael A.; Montalvo, Welby; Yagita, Hideo; Allison, James P. (2010-03-02). "PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors". Proceedings of the National Academy of Sciences. 107 (9): 4275–4280. doi:10.1073/pnas.0915174107. ISSN 0027-8424. PMC 2840093. PMID 20160101.{{cite journal}}: CS1 maint: PMC format (link)
  8. ^ Topalian, Suzanne L.; Taube, Janis M.; Anders, Robert A.; Pardoll, Drew M. "Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy". Nature Reviews. Cancer. 16 (5): 275–287. doi:10.1038/nrc.2016.36. ISSN 1474-1768. PMID 27079802.
  9. ^ Topalian, Suzanne L.; Taube, Janis M.; Anders, Robert A.; Pardoll, Drew M. "Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy". Nature Reviews. Cancer. 16 (5): 275–287. doi:10.1038/nrc.2016.36. ISSN 1474-1768. PMID 27079802.
  10. ^ Chen, Daniel S.; Mellman, Ira (2013-07-25). "Oncology meets immunology: the cancer-immunity cycle". Immunity. 39 (1): 1–10. doi:10.1016/j.immuni.2013.07.012. ISSN 1097-4180. PMID 23890059.
  11. ^ Topalian, Suzanne L.; Taube, Janis M.; Anders, Robert A.; Pardoll, Drew M. "Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy". Nature Reviews. Cancer. 16 (5): 275–287. doi:10.1038/nrc.2016.36. ISSN 1474-1768. PMID 27079802.
  12. ^ Topalian, Suzanne L.; Taube, Janis M.; Anders, Robert A.; Pardoll, Drew M. "Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy". Nature Reviews. Cancer. 16 (5): 275–287. doi:10.1038/nrc.2016.36. ISSN 1474-1768. PMID 27079802.
  13. ^ Porichis, Filippos; Kaufmann, Daniel E. (2017-02-26). "Role of PD-1 in HIV Pathogenesis and as Target for Therapy". Current HIV/AIDS reports. 9 (1): 81–90. doi:10.1007/s11904-011-0106-4. ISSN 1548-3568. PMC 3731769. PMID 22198819.{{cite journal}}: CS1 maint: PMC format (link)
  14. ^ Chew, Glen M.; Fujita, Tsuyoshi; Webb, Gabriela M.; Burwitz, Benjamin J.; Wu, Helen L.; Reed, Jason S.; Hammond, Katherine B.; Clayton, Kiera L.; Ishii, Naoto (2016-01-07). "TIGIT Marks Exhausted T Cells, Correlates with Disease Progression, and Serves as a Target for Immune Restoration in HIV and SIV Infection". PLoS Pathogens. 12 (1). doi:10.1371/journal.ppat.1005349. ISSN 1553-7366. PMC 4704737. PMID 26741490.{{cite journal}}: CS1 maint: PMC format (link) CS1 maint: unflagged free DOI (link)
  15. ^ Porichis, Filippos; Kaufmann, Daniel E. (2017-02-10). "Role of PD-1 in HIV Pathogenesis and as Target for Therapy". Current HIV/AIDS reports. 9 (1): 81–90. doi:10.1007/s11904-011-0106-4. ISSN 1548-3568. PMC 3731769. PMID 22198819.{{cite journal}}: CS1 maint: PMC format (link)
  16. ^ Chew, Glen M.; Fujita, Tsuyoshi; Webb, Gabriela M.; Burwitz, Benjamin J.; Wu, Helen L.; Reed, Jason S.; Hammond, Katherine B.; Clayton, Kiera L.; Ishii, Naoto (2016-01-07). "TIGIT Marks Exhausted T Cells, Correlates with Disease Progression, and Serves as a Target for Immune Restoration in HIV and SIV Infection". PLoS Pathogens. 12 (1). doi:10.1371/journal.ppat.1005349. ISSN 1553-7366. PMC 4704737. PMID 26741490.{{cite journal}}: CS1 maint: PMC format (link) CS1 maint: unflagged free DOI (link)
  17. ^ Kurnellas, Michael P.; Adams, Chris M.; Sobel, Raymond A.; Steinman, Lawrence; Rothbard, Jonathan B. (2013-04-03). "Amyloid Fibrils Composed of Hexameric Peptides Attenuate Neuroinflammation". Science translational medicine. 5 (179): 179ra42. doi:10.1126/scitranslmed.3005681. ISSN 1946-6234. PMC 3684024. PMID 23552370.{{cite journal}}: CS1 maint: PMC format (link)
  18. ^ Kurnellas, Michael Phillip; Ghosn, Eliver Eid Bou; Schartner, Jill M.; Baker, Jeanette; Rothbard, Jesse J.; Negrin, Robert S.; Herzenberg, Leonore A.; Fathman, C. Garrison; Steinman, Lawrence (2015-12-08). "Amyloid fibrils activate B-1a lymphocytes to ameliorate inflammatory brain disease". Proceedings of the National Academy of Sciences of the United States of America. 112 (49): 15016–15023. doi:10.1073/pnas.1521206112. ISSN 1091-6490. PMC 4679000. PMID 26621719.{{cite journal}}: CS1 maint: PMC format (link)
  19. ^ Kurnellas, Michael P.; Schartner, Jill M.; Fathman, C. Garrison; Jagger, Ann; Steinman, Lawrence; Rothbard, Jonathan B. (2014-08-25). "Mechanisms of action of therapeutic amyloidogenic hexapeptides in amelioration of inflammatory brain disease". Journal of Experimental Medicine. 211 (9): 1847–1856. doi:10.1084/jem.20140107. ISSN 0022-1007. PMC 4144739. PMID 25073790.{{cite journal}}: CS1 maint: PMC format (link)
  20. ^ Baruch, Kuti; Deczkowska, Aleksandra; Rosenzweig, Neta; Tsitsou-Kampeli, Afroditi; Sharif, Alaa Mohammad; Matcovitch-Natan, Orit; Kertser, Alexander; David, Eyal; Amit, Ido (2016-02-01). "PD-1 immune checkpoint blockade reduces pathology and improves memory in mouse models of Alzheimer's disease". Nature Medicine. 22 (2): 135–137. doi:10.1038/nm.4022. ISSN 1078-8956.
  21. ^ Saresella, Marina; Calabrese, Elena; Marventano, Ivana; Piancone, Federica; Gatti, Andrea; Farina, Elisabetta; Alberoni, Margherita; Clerici, Mario (2012-03-01). "A potential role for the PD1/PD-L1 pathway in the neuroinflammation of Alzheimer's disease". Neurobiology of Aging. 33 (3): 624.e11–22. doi:10.1016/j.neurobiolaging.2011.03.004. ISSN 1558-1497. PMID 21514692.