Immunotherapy

You might also like

Download as docx, pdf, or txt
Download as docx, pdf, or txt
You are on page 1of 17

Immunotherapy

From Wikipedia, the free encyclopedia

Jump to navigationJump to search

For the academic journal, see Immunotherapy (journal).

This article needs more medical references for verification or relies too heavily on primary sources.
Please review the contents of the article and add the appropriate references if you can. Unsourced or
poorly sourced material may be challenged and removed.

Find sources: "Immunotherapy" – news · newspapers · books · scholar · JSTOR (April 2018)

Rod of Asclepius2.svg

Question book-new.svg

This article relies too much on references to primary sources. Please improve this by adding secondary or
tertiary sources. (April 2018) (Learn how and when to remove this template message)

Immunotherapy

CAR T-cell Therapy.svg

The diagram above represents the process of chimeric antigen receptor T-cell therapy (CAR), this is a
method of immunotherapy, which is a growing practice in the treatment of cancer. The final result
should be a production of equipped T-cells that can recognize and fight the infected cancer cells in the
body.

T-cells (represented by objects labeled as ’t’) are removed from the patient's blood.

Then in a lab setting the gene that encodes for the specific antigen receptors are incorporated into the T-
cells.

Thus producing the CAR receptors (labeled as c) on the surface of the cells.

The newly modified T-cells are then further harvested and grown in the lab.

After a certain time period, the engineered T-cells are infused back into the patient.

MeSH D007167

OPS-301 code 8-03

[edit on Wikidata]

Immunotherapy or biological therapy is the treatment of disease by activating or suppressing the


immune system. Immunotherapies designed to elicit or amplify an immune response are classified as
activation immunotherapies, while immunotherapies that reduce or suppress are classified as
suppression immunotherapies.

In recent years, immunotherapy has become of great interest to researchers, clinicians and
pharmaceutical companies, particularly in its promise to treat various forms of cancer.[1][2][3]

Immunomodulatory regimens often have fewer side effects than existing drugs, including less potential
for creating resistance when treating microbial disease.[4]

Cell-based immunotherapies are effective for some cancers. Immune effector cells such as lymphocytes,
macrophages, dendritic cells, natural killer cells (NK Cell), cytotoxic T lymphocytes (CTL), etc., work
together to defend the body against cancer by targeting abnormal antigens expressed on the surface of
tumor cells.

Therapies such as granulocyte colony-stimulating factor (G-CSF), interferons, imiquimod and cellular
membrane fractions from bacteria are licensed for medical use. Others including IL-2, IL-7, IL-12, various
chemokines, synthetic cytosine phosphate-guanosine (CpG) oligodeoxynucleotides and glucans are
involved in clinical and preclinical studies.

Contents

1 Immunomodulators

2 Activation immunotherapies

2.1 Cancer

3 Immune enhancement therapy

4 Suppression immunotherapies

4.1 Immunosuppressive drugs

4.2 Immune tolerance

4.3 Allergies

5 Helminthic therapies

6 See also

7 References
8 External links

Immunomodulators

Immunomodulators are the active agents of immunotherapy. They are a diverse array of recombinant,
synthetic, and natural preparations.

Class Example agents

Interleukins IL-2, IL-7, IL-12

Cytokines Interferons, G-CSF

Chemokines CCL3, CCL26, CXCL7

Immunomodulatory imide drugs (IMiDs) thalidomide and its analogues (lenalidomide,


pomalidomide, and apremilast)

Other cytosine phosphate-guanosine, oligodeoxynucleotides, glucans

Activation immunotherapies

Cancer

Main article: Cancer immunotherapy

Cancer treatment used to be focused on killing or removing cancer cells and tumors, with chemotherapy
or surgery or radiation. These treatments can be very effective and in many cases are still used. In 2018
the Nobel Prize in Physiology or Medicine was awarded to James P. Allison and Tasuku Honjo “for their
discovery of cancer therapy by inhibition of negative immune regulation.” Cancer immunotherapy
attempts to stimulate the immune system to destroy tumors. A variety of strategies are in use or are
undergoing research and testing. Randomized controlled studies in different cancers resulting in
significant increase in survival and disease free period have been reported[2] and its efficacy is enhanced
by 20–30% when cell-based immunotherapy is combined with conventional treatment methods.[2]

One of the oldest forms of cancer immunotherapy is the use of BCG vaccine, which was originally to
vaccinate against tuberculosis and later was found to be useful in the treatment of bladder cancer.[5]
BCG immunotherapy induces both local and systemic immune responses. The mechanisms by which BCG
immunotherapy mediates tumour immunity have been widely studied, but they are still not completely
understood.[6]

The use of monoclonal antibodies in cancer therapy was first introduced in 1997 with rituximab, an anti-
CD20 antibody for treatment of B cell lymphoma.[7] Since then several monoclonal antibodies have been
approved for treatment of various hematological malignancies as well as for solid tumors.[8][9]
The extraction of G-CSF lymphocytes from the blood and expanding in vitro against a tumour antigen
before reinjecting the cells with appropriate stimulatory cytokines. The cells then destroy the tumor cells
that express the antigen.[citation needed]Topical immunotherapy utilizes an immune enhancement
cream (imiquimod) which produces interferon, causing the recipient's killer T cells to destroy warts,[10]
actinic keratoses, basal cell cancer, vaginal intraepithelial neoplasia,[11] squamous cell cancer,[12][13]
cutaneous lymphoma,[14] and superficial malignant melanoma.[15] Injection immunotherapy
("intralesional" or "intratumoral") uses mumps, candida, the HPV vaccine[16][17] or trichophytin antigen
injections to treat warts (HPV induced tumors).

Adoptive cell transfer has been tested on lung [18] and other cancers, with greatest success achieved in
melanoma.

Dendritic cell-based pump-priming or vaccination

Dendritic cells (DC) can be stimulated to activate a cytotoxic response towards an antigen. Dendritic
cells, a type of antigen-presenting cell, are harvested from the person needing the immunotherapy.
These cells are then either pulsed with an antigen or tumor lysate or transfected with a viral vector,
causing them to display the antigen. Upon transfusion into the person, these activated cells present the
antigen to the effector lymphocytes (CD4+ helper T cells, cytotoxic CD8+ T cells and B cells). This initiates
a cytotoxic response against tumor cells expressing the antigen (against which the adaptive response has
now been primed). The cancer vaccine Sipuleucel-T is one example of this approach.[19]

The current approaches for DC-based vaccination are mainly based on antigen loading on in vitro-
generated DCs from monocytes or CD34+ cells, activating them with different TLR ligands, cytokine
combinations, and injecting them back to the patients. The in vivo targeting approaches comprise
administering specific cytokines (e.g., Flt3L, GM-CSF) and targeting the DCs with antibodies to C-type
lectin receptors or agonistic antibodies (e.g., anti-CD40) that are conjugated with antigen of interest.
Future approach may target DC subsets based on their specifically expressed C-type lectin receptors or
chemokine receptors. Another potential approach is the generation of genetically engineered DCs from
induced pluripotent stem cells and use of neoantigen-loaded DCs for inducing better clinical outcome.
[20]

T-cell adoptive transfer

Adoptive cell transfer in vitro cultivates autologous, extracted T cells for later transfusion.[21]

Alternatively, Genetically engineered T cells are created by harvesting T cells and then infecting the T
cells with a retrovirus that contains a copy of a T cell receptor (TCR) gene that is specialised to recognise
tumour antigens. The virus integrates the receptor into the T cells' genome. The cells are expanded non-
specifically and/or stimulated. The cells are then reinfused and produce an immune response against the
tumour cells.[22] The technique has been tested on refractory stage IV metastatic melanomas[21] and
advanced skin cancer.[23][24][25]

Whether T cells are genetically engineered or not, before reinfusion, lymphodepletion of the recipient is
required to eliminate regulatory T cells as well as unmodified, endogenous lymphocytes that compete
with the transferred cells for homeostatic cytokines.[21][26][27][28] Lymphodepletion may be achieved
by myeloablative chemotherapy, to which total body irradiation may be added for greater effect.[29]
Transferred cells multiplied in vivo and persisted in peripheral blood in many people, sometimes
representing levels of 75% of all CD8+ T cells at 6–12 months after infusion.[30] As of 2012, clinical trials
for metastatic melanoma were ongoing at multiple sites.[31] Clinical responses to adoptive transfer of T
cells were observed in patients with metastatic melanoma resistant to multiple immunotherapies.[32]

Checkpoint inhibitors

Main article: Checkpoint inhibitor

Anti-PD-1/PD-L1 and anti-CTLA-4 antibodies are the two types of checkpoint inhibitors currently
available to patients.The approval of anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and anti-
programmed cell death protein 1 (PD-1) antibodies for human use has already resulted in significant
improvements in disease outcomes for various cancers.[33]

Although these molecules were originally discovered as molecules playing a role in T cell activation or
apoptosis, subsequent preclinical research showed their important role in the maintenance of peripheral
immune tolerance.[34]

Immune checkpoint inhibitors are approved to treat some patients with a variety of cancer types,
including melanoma, breast cancer, bladder cancer, cervical cancer, colon cancer, head and neck cancer,
or hodgkin lymphoma.[35]

These therapies have revolutionized cancer immunotherapy as they showed for the first time in many
years of research in metastatic melanoma, which is considered one of the most immunogenic human
cancers, an improvement in overall survival, with an increasing group of patients benefitting long-term
from these treatments.[34]

Immune enhancement therapy

Autologous immune enhancement therapy use a person's own peripheral blood-derived natural killer
cells, cytotoxic T lymphocytes, epithelial cells and other relevant immune cells are expanded in vitro and
then reinfused.[36] The therapy has been tested against Hepatitis C,[37][38][39] Chronic fatigue
syndrome[40][41] and HHV6 infection.[42]

Suppression immunotherapies

Immune suppression dampens an abnormal immune response in autoimmune diseases or reduces a


normal immune response to prevent rejection of transplanted organs or cells.

Immunosuppressive drugs

Immunosuppressive drugs help manage organ transplantation and autoimmune disease. Immune
responses depend on lymphocyte proliferation. Cytostatic drugs are immunosuppressive.
Glucocorticoids are somewhat more specific inhibitors of lymphocyte activation, whereas inhibitors of
immunophilins more specifically target T lymphocyte activation. Immunosuppressive antibodies target
steps in the immune response. Other drugs modulate immune responses and can be used to induce
immune regulation. It has been observed in a preclinical trial that regulation of the immune system by
small immunosuppressive molecules such as Vitamin D and Dexamethasone, administered under a low-
dose regimen and subcutaneously, could be helpful in preventing or treating chronic inflammation.[43]

Immune tolerance

The body naturally does not launch an immune system attack on its own tissues. Models generally
identify CD4+ T-cells at the centre of the autoimmune response. Loss of T-cell tolerance then unleashes
B-cells and other immune effector cells on to the target tissue. The ideal tolerogenic therapy would
target the specific T-cell clone(s) co-ordinating the autoimmune attack.[44]

Immune tolerance therapies seek to reset the immune system so that the body stops mistakenly
attacking its own organs or cells in autoimmune disease or accepts foreign tissue in organ
transplantation.[45] A recent therapeutic approach is the infusion of regulatory immune cells into
transplant recipients. The transfer of regulatory immune cells has the potential to inhibit the activity of
effector.[46][47]

Creating immune tolerance reduces or eliminates the need for lifelong immunosuppression and
attendant side effects. It has been tested on transplantations, rehumatoid arthritis, type 1 diabetes and
other autoimmune disorders.

Approaches to therapeutic tolerance induction[44][48][49]


Modality Details

Non-antigen specific • Monoclonal Antibodies Depleting

Anti-CD52

Anti-CD4

Anti-LFA2

Non-depleting

Anti-CD4

Anti-CD3

Anti-LFA-1

CTLA4-Ig

Anti-CD25

• Haematopoietic stem cell transplantation Non-myeloablative Myeloablative

• Mesenchymal stem cell transplantation

• Regulatory T cell therapy Non-antigen specific Antigen-specific

• Low dose IL-2 to expand regulatory T cells

• Microbiome manipulation

Antigen specific• Peptide therapy Subcutaneous, intradermal, Mucosal (oral, inhaled)

Tolerogenic dendritic cells, liposomes and nanoparticles


• Altered peptide ligands

Allergies

Main article: Allergen immunotherapy

Immunotherapy can also be used to treat allergies. While allergy treatments (such as antihistamines or
corticosteroids) treat allergic symptoms, immunotherapy can reduce sensitivity to allergens, lessening its
severity.

Immunotherapy may produce long-term benefits.[50] Immunotherapy is partly effective in some people
and ineffective in others, but it offers allergy sufferers a chance to reduce or stop their symptoms.

The therapy is indicated for people who are extremely allergic or who cannot avoid specific allergens.

IgE-mediated food allergy is a global health problem that affects millions of persons and affects every
aspect of life for the patient.[51] A promising approach to treat food allergies is the use of oral
immunotherapy (OIT). OIT consists in a gradual exposure to increasing amounts of allergen can lead to
the majority of subjects tolerating doses of food sufficient to prevent reaction on accidental exposure.
[52] Dosages increase over time, as the person becomes desensitized. This technique has been tested on
infants to prevent peanut allergies.[53]

Allergen-specific immunotherapy (ASIT) has become the gold standard for the causative treatment for
IgE‐mediated allergic diseases for a large variety of allergens. One may curiously await the new
developments, which will further enhance our understanding of allergy mechanisms and improve ASIT
for the next generations of patients and physicians.[54]

Helminthic therapies

Whipworm ova (Trichuris suis) and Hookworm (Necator americanus) have been tested for immunological
diseases and allergies. Helminthic therapy has been investigated as a treatment for relapsing remitting
multiple sclerosis[55] Crohn's,[56][57][58] allergies and asthma.[59] The mechanism of how the
helminths modulate the immune response, is unknown. Hypothesized mechanisms include re-
polarisation of the Th1 / Th2 response[60] and modulation of dendritic cell function.[61][62] The
helminths down regulate the pro-inflammatory Th1 cytokines, Interleukin-12 (IL-12), Interferon-Gamma
(IFN-γ) and Tumour Necrosis Factor-Alpha (TNF-ά), while promoting the production of regulatory Th2
cytokines such as IL-10, IL-4, IL-5 and IL-13.[60][63]

Co-evolution with helminths has shaped some of the genes associated with Interleukin expression and
immunological disorders, such Crohn's, ulcerative colitis and celiac disease. Helminth's relationship to
humans as hosts should be classified as mutualistic or symbiotic.[citation needed]

See also

Biological response modifier

Sepsivac

Checkpoint inhibitor

Interleukin-2 immunotherapy

Immunostimulant

Microtransplantation

Photoimmunotherapy in vitro or in vivo[64][65][66][67][68][69][70]

References

"Immunotherapy | Memorial Sloan Kettering Cancer Center". mskcc.org. Retrieved 2017-07-27.

Syn NL, Teng MW, Mok TS, Soo RA (December 2017). "De-novo and acquired resistance to immune
checkpoint targeting". The Lancet. Oncology. 18 (12): e731–e741. doi:10.1016/s1470-2045(17)30607-1.
PMID 29208439.

Conforti L (February 2012). "The ion channel network in T lymphocytes, a target for immunotherapy".
Clinical Immunology. 142 (2): 105–6. doi:10.1016/j.clim.2011.11.009. PMID 22189042.

Masihi KN (July 2001). "Fighting infection using immunomodulatory agents". Expert Opinion on
Biological Therapy. 1 (4): 641–53. doi:10.1517/14712598.1.4.641. PMID 11727500. S2CID 33308465.

Fuge O, Vasdev N, Allchorne P, Green JS (2015). "Immunotherapy for bladder cancer". Research and
Reports in Urology. 7: 65–79. doi:10.2147/RRU.S63447. PMC 4427258. PMID 26000263.

Pettenati, Caroline; Ingersoll, Molly A. (October 2018). "Mechanisms of BCG immunotherapy and its
outlook for bladder cancer". Nature Reviews Urology. 15 (10): 615–625. doi:10.1038/s41585-018-0055-4.
ISSN 1759-4820. PMID 29991725. S2CID 49670901.

Salles G, Barrett M, Foà R, Maurer J, O'Brien S, Valente N, et al. (October 2017). "Rituximab in B-Cell
Hematologic Malignancies: A Review of 20 Years of Clinical Experience". Advances in Therapy. 34 (10):
2232–2273. doi:10.1007/s12325-017-0612-x. PMC 5656728. PMID 28983798.

Hoos A (April 2016). "Development of immuno-oncology drugs - from CTLA4 to PD1 to the next
generations". Nature Reviews. Drug Discovery. 15 (4): 235–47. doi:10.1038/nrd.2015.35. PMID
26965203. S2CID 54550859.

Pento JT (November 2017). "Monoclonal Antibodies for the Treatment of Cancer". Anticancer Research.
37 (11): 5935–5939. doi:10.21873/anticanres.12040. PMID 29061772.
van Seters M, van Beurden M, ten Kate FJ, Beckmann I, Ewing PC, Eijkemans MJ, et al. (April 2008).
"Treatment of vulvar intraepithelial neoplasia with topical imiquimod". The New England Journal of
Medicine. 358 (14): 1465–73. doi:10.1056/NEJMoa072685. PMID 18385498.

Buck HW, Guth KJ (October 2003). "Treatment of vaginal intraepithelial neoplasia (primarily low grade)
with imiquimod 5% cream". Journal of Lower Genital Tract Disease. 7 (4): 290–3. doi:10.1097/00128360-
200310000-00011. PMID 17051086. S2CID 44649376.

Järvinen R, Kaasinen E, Sankila A, Rintala E (August 2009). "Long-term efficacy of maintenance bacillus
Calmette-Guérin versus maintenance mitomycin C instillation therapy in frequently recurrent TaT1
tumours without carcinoma in situ: a subgroup analysis of the prospective, randomised FinnBladder I
study with a 20-year follow-up". European Urology. 56 (2): 260–5. doi:10.1016/j.eururo.2009.04.009.
PMID 19395154.

Davidson HC, Leibowitz MS, Lopez-Albaitero A, Ferris RL (September 2009). "Immunotherapy for head
and neck cancer". Oral Oncology. 45 (9): 747–51. doi:10.1016/j.oraloncology.2009.02.009. PMID
19442565.

Dani T, Knobler R (January 2009). "Extracorporeal photoimmunotherapy-photopheresis". Frontiers in


Bioscience. 14 (14): 4769–77. doi:10.2741/3566. PMID 19273388.

Eggermont AM, Schadendorf D (June 2009). "Melanoma and immunotherapy". Hematology/Oncology


Clinics of North America. 23 (3): 547–64, ix–x. doi:10.1016/j.hoc.2009.03.009. PMID 19464602.

Chuang CM, Monie A, Wu A, Hung CF (May 2009). "Combination of apigenin treatment with therapeutic
HPV DNA vaccination generates enhanced therapeutic antitumor effects". Journal of Biomedical Science.
16 (1): 49. doi:10.1186/1423-0127-16-49. PMC 2705346. PMID 19473507.

Pawlita M, Gissmann L (April 2009). "[Recurrent respiratory papillomatosis: indication for HPV
vaccination?]". Deutsche Medizinische Wochenschrift (in German). 134 Suppl 2: S100-2. doi:10.1055/s-
0029-1220219. PMID 19353471.

Kang N, Zhou J, Zhang T, Wang L, Lu F, Cui Y, et al. (August 2009). "Adoptive immunotherapy of lung
cancer with immobilized anti-TCRgammadelta antibody-expanded human gammadelta T-cells in
peripheral blood". Cancer Biology & Therapy. 8 (16): 1540–9. doi:10.4161/cbt.8.16.8950. PMID
19471115.

Di Lorenzo G, Buonerba C, Kantoff PW (May 2011). "Immunotherapy for the treatment of prostate
cancer". Nature Reviews. Clinical Oncology. 8 (9): 551–61. doi:10.1038/nrclinonc.2011.72. PMID
21606971. S2CID 5337484.

Sabado, Rachel L; Balan, Sreekumar; Bhardwaj, Nina (January 2017). "Dendritic cell-based
immunotherapy". Cell Research. 27 (1): 74–95. doi:10.1038/cr.2016.157. ISSN 1001-0602. PMC 5223236.
PMID 28025976.

Rosenberg SA, Restifo NP, Yang JC, Morgan RA, Dudley ME (April 2008). "Adoptive cell transfer: a clinical
path to effective cancer immunotherapy". Nature Reviews. Cancer. 8 (4): 299–308. doi:10.1038/nrc2355.
PMC 2553205. PMID 18354418.

Morgan RA, Dudley ME, Wunderlich JR, Hughes MS, Yang JC, Sherry RM, et al. (October 2006). "Cancer
regression in patients after transfer of genetically engineered lymphocytes". Science. 314 (5796): 126–9.
Bibcode:2006Sci...314..126M. doi:10.1126/science.1129003. PMC 2267026. PMID 16946036.

Hunder NN, Wallen H, Cao J, Hendricks DW, Reilly JZ, Rodmyre R, et al. (June 2008). "Treatment of
metastatic melanoma with autologous CD4+ T cells against NY-ESO-1". The New England Journal of
Medicine. 358 (25): 2698–703. doi:10.1056/NEJMoa0800251. PMC 3277288. PMID 18565862.

"2008 Symposium Program & Speakers". Cancer Research Institute. Archived from the original on 2008-
10-15.

Highfield R (18 June 2008). "Cancer patient recovers after injection of immune cells". The Telegraph.
Archived from the original on 12 September 2008. Retrieved 22 December 2019.

Antony PA, Piccirillo CA, Akpinarli A, Finkelstein SE, Speiss PJ, Surman DR, et al. (March 2005). "CD8+ T
cell immunity against a tumor/self-antigen is augmented by CD4+ T helper cells and hindered by
naturally occurring T regulatory cells". Journal of Immunology. 174 (5): 2591–601.
doi:10.4049/jimmunol.174.5.2591. PMC 1403291. PMID 15728465.

Gattinoni L, Finkelstein SE, Klebanoff CA, Antony PA, Palmer DC, Spiess PJ, et al. (October 2005).
"Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively
transferred tumor-specific CD8+ T cells". The Journal of Experimental Medicine. 202 (7): 907–12.
doi:10.1084/jem.20050732. PMC 1397916. PMID 16203864.

Dummer W, Niethammer AG, Baccala R, Lawson BR, Wagner N, Reisfeld RA, Theofilopoulos AN (July
2002). "T cell homeostatic proliferation elicits effective antitumor autoimmunity". The Journal of Clinical
Investigation. 110 (2): 185–92. doi:10.1172/JCI15175. PMC 151053. PMID 12122110.

Dudley ME, Yang JC, Sherry R, Hughes MS, Royal R, Kammula U, et al. (November 2008). "Adoptive cell
therapy for patients with metastatic melanoma: evaluation of intensive myeloablative chemoradiation
preparative regimens". Journal of Clinical Oncology. 26 (32): 5233–9. doi:10.1200/JCO.2008.16.5449.
PMC 2652090. PMID 18809613.

Dudley ME, Wunderlich JR, Robbins PF, Yang JC, Hwu P, Schwartzentruber DJ, et al. (October 2002).
"Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes".
Science. 298 (5594): 850–4. Bibcode:2002Sci...298..850D. doi:10.1126/science.1076514. PMC 1764179.
PMID 12242449.

Pilon-Thomas S, Kuhn L, Ellwanger S, Janssen W, Royster E, Marzban S, et al. (October 2012). "Efficacy of
adoptive cell transfer of tumor-infiltrating lymphocytes after lymphopenia induction for metastatic
melanoma". Journal of Immunotherapy. 35 (8): 615–20. doi:10.1097/CJI.0b013e31826e8f5f. PMC
4467830. PMID 22996367.

Andersen R, Borch TH, Draghi A, Gokuldass A, Rana MA, Pedersen M, et al. (July 2018). "T cells isolated
from patients with checkpoint inhibitor-resistant melanoma are functional and can mediate tumor
regression". Annals of Oncology. 29 (7): 1575–1581. doi:10.1093/annonc/mdy139. PMID 29688262.

Seidel, Judith A.; Otsuka, Atsushi; Kabashima, Kenji (2018-03-28). "Anti-PD-1 and Anti-CTLA-4 Therapies
in Cancer: Mechanisms of Action, Efficacy, and Limitations". Frontiers in Oncology. 8: 86.
doi:10.3389/fonc.2018.00086. ISSN 2234-943X. PMC 5883082. PMID 29644214.
Haanen, John B. A. G.; Robert, Caroline (2015). "Immune Checkpoint Inhibitors". Immuno-Oncology.
Progress in Tumor Research. 42: 55–66. doi:10.1159/000437178. ISBN 978-3-318-05589-4. PMID
26382943.

"Immune Checkpoint Inhibitors - National Cancer Institute". www.cancer.gov. 2019-09-24. Retrieved


2020-08-24.

Manjunath SR, Ramanan G, Dedeepiya VD, Terunuma H, Deng X, Baskar S, et al. (January 2012).
"Autologous immune enhancement therapy in recurrent ovarian cancer with metastases: a case report".
Case Reports in Oncology. 5 (1): 114–8. doi:10.1159/000337319. PMC 3364094. PMID 22666198.

Li Y, Zhang T, Ho C, Orange JS, Douglas SD, Ho WZ (December 2004). "Natural killer cells inhibit hepatitis
C virus expression". Journal of Leukocyte Biology. 76 (6): 1171–9. doi:10.1189/jlb.0604372. PMID
15339939.

Doskali M, Tanaka Y, Ohira M, Ishiyama K, Tashiro H, Chayama K, Ohdan H (March 2011). "Possibility of
adoptive immunotherapy with peripheral blood-derived CD3⁻CD56+ and CD3+CD56+ cells for inducing
antihepatocellular carcinoma and antihepatitis C virus activity". Journal of Immunotherapy. 34 (2): 129–
38. doi:10.1097/CJI.0b013e3182048c4e. PMID 21304407. S2CID 26385818.

Terunuma H, Deng X, Dewan Z, Fujimoto S, Yamamoto N (2008). "Potential role of NK cells in the
induction of immune responses: implications for NK cell-based immunotherapy for cancers and viral
infections". International Reviews of Immunology. 27 (3): 93–110. doi:10.1080/08830180801911743.
PMID 18437601. S2CID 27557213.

See DM, Tilles JG (1996). "alpha-Interferon treatment of patients with chronic fatigue syndrome".
Immunological Investigations. 25 (1–2): 153–64. doi:10.3109/08820139609059298. PMID 8675231.

Ojo-Amaize EA, Conley EJ, Peter JB (January 1994). "Decreased natural killer cell activity is associated
with severity of chronic fatigue immune dysfunction syndrome". Clinical Infectious Diseases. 18 Suppl 1:
S157-9. doi:10.1093/clinids/18.Supplement_1.S157. PMID 8148445.

Kida K, Isozumi R, Ito M (December 2000). "Killing of human Herpes virus 6-infected cells by
lymphocytes cultured with interleukin-2 or -12". Pediatrics International. 42 (6): 631–6.
doi:10.1046/j.1442-200x.2000.01315.x. PMID 11192519.

Ospina-Quintero, Laura; Jaramillo, Julio C.; Tabares-Guevara, Jorge H.; Ramírez-Pineda, José R. (24 April
2020). "Reformulating Small Molecules for Cardiovascular Disease Immune Intervention: Low-Dose
Combined Vitamin D/Dexamethasone Promotes IL-10 Production and Atheroprotection in Dyslipidemic
Mice". Frontiers in Immunology. 11: 743. doi:10.3389/fimmu.2020.00743. PMC 7197409. PMID
32395119.

Rayner, Fiona; Isaacs, John D (2018-12-01). "Therapeutic tolerance in autoimmune disease". Seminars in
Arthritis and Rheumatism. 48 (3): 558–562. doi:10.1016/j.semarthrit.2018.09.008. ISSN 0049-0172.
PMID 30348449.

Rotrosen D, Matthews JB, Bluestone JA (July 2002). "The immune tolerance network: a new paradigm
for developing tolerance-inducing therapies". The Journal of Allergy and Clinical Immunology. 110 (1):
17–23. doi:10.1067/mai.2002.124258. PMID 12110811. S2CID 30884739.
Stolp, Jessica; Zaitsu, Masaaki; Wood, Kathryn J. (2019), Boyd, Ashleigh S. (ed.), "Immune Tolerance and
Rejection in Organ Transplantation", Immunological Tolerance, New York, NY: Springer New York, 1899,
pp. 159–180, doi:10.1007/978-1-4939-8938-6_12, ISBN 978-1-4939-8936-2, PMID 30649772

McMurchy, Alicia N.; Bushell, Andrew; Levings, Megan K.; Wood, Kathryn J. (2011-08-01). "Moving to
tolerance: Clinical application of T regulatory cells". Seminars in Immunology. Advances in
Transplantation. 23 (4): 304–313. doi:10.1016/j.smim.2011.04.001. ISSN 1044-5323. PMC 3836227.
PMID 21620722.

Baker, Kenneth F; Isaacs, John D (March 2014). "Prospects for Therapeutic Tolerance in Humans".
Current Opinion in Rheumatology. 26 (2): 219–227. doi:10.1097/BOR.0000000000000029. ISSN 1040-
8711. PMC 4640179. PMID 24378931.

Cooles, Faye A. H.; Isaacs, John D. (2010-08-01). "Treating to re-establish tolerance in inflammatory
arthritis – lessons from other diseases". Best Practice & Research Clinical Rheumatology.
Pharmacotherapy: Concepts of Pathogenesis and Emerging Treatments. 24 (4): 497–511.
doi:10.1016/j.berh.2010.01.007. ISSN 1521-6942. PMID 20732648.

Durham SR, Walker SM, Varga EM, Jacobson MR, O'Brien F, Noble W, et al. (August 1999). "Long-term
clinical efficacy of grass-pollen immunotherapy". The New England Journal of Medicine. 341 (7): 468–75.
doi:10.1056/NEJM199908123410702. PMID 10441602. S2CID 14629112.

Jones, Stacie M.; Burks, A. Wesley; Dupont, Christophe (2014-02-01). "State of the art on food allergen
immunotherapy: Oral, sublingual, and epicutaneous". Journal of Allergy and Clinical Immunology. 133
(2): 318–323. doi:10.1016/j.jaci.2013.12.1040. ISSN 0091-6749. PMID 24636471.

MacGinnitie, Andrew J.; Rachid, Rima; Gragg, Hana; Little, Sara V.; Lakin, Paul; Cianferoni, Antonella;
Heimall, Jennifer; Makhija, Melanie; Robison, Rachel; Chinthrajah, R. Sharon; Lee, John (2017-03-01).
"Omalizumab facilitates rapid oral desensitization for peanut allergy". Journal of Allergy and Clinical
Immunology. 139 (3): 873–881.e8. doi:10.1016/j.jaci.2016.08.010. ISSN 0091-6749. PMC 5369605. PMID
27609658. S2CID 3626708.

"Clinical Trials Search Results - Stanford University School of Medicine". med.stanford.edu. Retrieved
2016-04-03.

Ring, J.; Gutermuth, J. (2011). "100 years of hyposensitization: history of allergen-specific


immunotherapy (ASIT)". Allergy. 66 (6): 713–724. doi:10.1111/j.1398-9995.2010.02541.x. ISSN 1398-
9995. PMID 21320133. S2CID 19390006.

Correale J, Farez M (February 2007). "Association between parasite infection and immune responses in
multiple sclerosis". Annals of Neurology. 61 (2): 97–108. doi:10.1002/ana.21067. PMID 17230481. S2CID
1033417.

Croese J, O'neil J, Masson J, Cooke S, Melrose W, Pritchard D, Speare R (January 2006). "A proof of
concept study establishing Necator americanus in Crohn's patients and reservoir donors". Gut. 55 (1):
136–7. doi:10.1136/gut.2005.079129. PMC 1856386. PMID 16344586.

Reddy A, Fried B (January 2009). "An update on the use of helminths to treat Crohn's and other
autoimmunune diseases". Parasitology Research. 104 (2): 217–21. doi:10.1007/s00436-008-1297-5.
PMID 19050918. S2CID 19279688.

Laclotte C, Oussalah A, Rey P, Bensenane M, Pluvinage N, Chevaux JB, et al. (December 2008).
"[Helminths and inflammatory bowel diseases]". Gastroenterologie Clinique et Biologique (in French). 32
(12): 1064–74. doi:10.1016/j.gcb.2008.04.030. PMID 18619749.

Zaccone P, Fehervari Z, Phillips JM, Dunne DW, Cooke A (October 2006). "Parasitic worms and
inflammatory diseases". Parasite Immunology. 28 (10): 515–23. doi:10.1111/j.1365-3024.2006.00879.x.
PMC 1618732. PMID 16965287.

Brooker S, Bethony J, Hotez PJ (2004). Human Hookworm Infection in the 21st Century. Advances in
Parasitology. 58. pp. 197–288. doi:10.1016/S0065-308X(04)58004-1. ISBN 9780120317585. PMC
2268732. PMID 15603764.

Fujiwara RT, Cançado GG, Freitas PA, Santiago HC, Massara CL, Dos Santos Carvalho O, et al. (2009).
Yazdanbakhsh M (ed.). "Necator americanus infection: a possible cause of altered dendritic cell
differentiation and eosinophil profile in chronically infected individuals". PLOS Neglected Tropical
Diseases. 3 (3): e399. doi:10.1371/journal.pntd.0000399. PMC 2654967. PMID 19308259.

Carvalho L, Sun J, Kane C, Marshall F, Krawczyk C, Pearce EJ (January 2009). "Review series on helminths,
immune modulation and the hygiene hypothesis: mechanisms underlying helminth modulation of
dendritic cell function". Immunology. 126 (1): 28–34. doi:10.1111/j.1365-2567.2008.03008.x. PMC
2632707. PMID 19120496.

Fumagalli M, Pozzoli U, Cagliani R, Comi GP, Riva S, Clerici M, et al. (June 2009). "Parasites represent a
major selective force for interleukin genes and shape the genetic predisposition to autoimmune
conditions". The Journal of Experimental Medicine. 206 (6): 1395–408. doi:10.1084/jem.20082779. PMC
2715056. PMID 19468064.

Hong CH, Tang MR, Hsu SH, Yang CH, Tseng CS, Ko YC, et al. (September 2019). "Enhanced early immune
response of leptospiral outer membrane protein LipL32 stimulated by narrow band mid-infrared
exposure". Journal of Photochemistry and Photobiology. B, Biology. 198: 111560.
doi:10.1016/j.jphotobiol.2019.111560. PMID 31336216.

Chang HY, Li MH, Huang TC, Hsu CL, Tsai SR, Lee SC, et al. (February 2015). "Quantitative proteomics
reveals middle infrared radiation-interfered networks in breast cancer cells". Journal of Proteome
Research. 14 (2): 1250–62. doi:10.1021/pr5011873. PMID 25556991.

Nagaya T, Okuyama S, Ogata F, Maruoka Y, Choyke PL, Kobayashi H (May 2019). "Near infrared
photoimmunotherapy using a fiber optic diffuser for treating peritoneal gastric cancer dissemination".
Gastric Cancer. 22 (3): 463–472. doi:10.1007/s10120-018-0871-5. PMC 7400986. PMID 30171392.

Mitsunaga M, Ogawa M, Kosaka N, Rosenblum LT, Choyke PL, Kobayashi H (November 2011). "Cancer
cell-selective in vivo near infrared photoimmunotherapy targeting specific membrane molecules".
Nature Medicine. 17 (12): 1685–91. doi:10.1038/nm.2554. PMC 3233641. PMID 22057348.

Sato K, Sato N, Xu B, Nakamura Y, Nagaya T, Choyke PL, et al. (August 2016). "Spatially selective
depletion of tumor-associated regulatory T cells with near-infrared photoimmunotherapy". Science
Translational Medicine. 8 (352): 352ra110. doi:10.1126/scitranslmed.aaf6843. PMID 27535621.
Nagaya T, Nakamura Y, Sato K, Harada T, Choyke PL, Kobayashi H (June 2016). "Improved micro-
distribution of antibody-photon absorber conjugates after initial near infrared photoimmunotherapy
(NIR-PIT)". Journal of Controlled Release. 232: 1–8. doi:10.1016/j.jconrel.2016.04.003. PMC 4893891.
PMID 27059723.

Zhen Z, Tang W, Wang M, Zhou S, Wang H, Wu Z, et al. (February 2017). "Protein Nanocage Mediated
Fibroblast-Activation Protein Targeted Photoimmunotherapy To Enhance Cytotoxic T Cell Infiltration and
Tumor Control". Nano Letters. 17 (2): 862–869. Bibcode:2017NanoL..17..862Z.
doi:10.1021/acs.nanolett.6b04150. PMID 28027646.

External links

Langreth R (12 February 2009). "Cancer Miracles". Forbes.

International Society for Biological Therapy of Cancer

Cancer Research Institute Annual International Cancer Immunotherapy Symposia Series

The story behind immunotherapy’s innovative cellular voyage

Issoria lathonia.jpgBiology portalWHO Rod.svgMedicine portal

Authority control Edit this at Wikidata

NDL: 00567589

Categories: ImmunotherapyCancer immunotherapyVirotherapy

Navigation menu

Not logged inTalkContributionsCreate accountLog in

ArticleTalk

ReadEditView historySearch

Search Wikipedia

Main page

Contents

Current events

Random article

About Wikipedia

Contact us

Donate

Contribute
Help

Learn to edit

Community portal

Recent changes

Upload file

Tools

What links here

Related changes

Special pages

Permanent link

Page information

Cite this page

Wikidata item

Print/export

Download as PDF

Printable version

In other projects

Wikimedia Commons

Languages

‫العربية‬
Deutsch

Español

Français

Bahasa Indonesia

Nederlands

Русский
‫اردو‬
中文

22 more

Edit links

This page was last edited on 23 September 2020, at 08:14 (UTC).

Text is available under the Creative Commons Attribution-ShareAlike License; additional terms may
apply. By using this site, you agree to the Terms of Use and Privacy Policy. Wikipedia® is a registered
trademark of the Wikimedia Fo

You might also like