Pharmacology of immunomodulating drugs Immune system : The importance of the immune system is to protect the body against harmful foreign molecules (living and non-living pathogens) and defend against diseases. Components of immune system: 1- Lymphoid organs and tissues 2- Leukocytes (WBCs) 3- Soluble proteins: Immune system (cont’d) 1- Lymphoid organs and tissues 2- Leukocytes - Granulocytes - Monocytes (Phagocytes) - Lymphocytes 3- Soluble proteins: - Complement system. - Acute phase proteins. - Antibodies. - Cytokines.(are soluble , signaling proteins that produced by various population of immune cells and other cells to interact with cell surface receptors on a variety of affecter cells and they play many roles as pro-inflammatory, anti-nflammatory, growth regulation and others). Cytokines include ◦ ◦ ◦ ◦ ◦ Interleukins (ILs) Interferons (IFNs), Tumor Necrosis Factors (TNFs), Transforming Growth Factors (TGFs) Colony-stimulating factors (CSFs). Mammalian Immunity 1- Innate immunity • • • Non- specific, constitutive. First line of defense. Present from birth. No immunologic memory. • Includes physical, chemical and biochemical barriers. • Includes physical, chemical and biochemical barriers. • Inflammation 2- Acquired Immunity • • • • • It is secondary to the innate response. Adaptive and specific. Recognition ( self/ non-self or pathogenic discrimination). Essential to the development of specific immunity is the recognition of antigen. Immunological memory (long lasting). • Two types of specific immune response: 1- Cell-mediated immunity (CMI): All immunologic activities in which cellular elements play a direct role e.g. activation of antigen-specific T-lymphocytes. 2- Humoral immunity (HI): It is directly dependent on the production of antigen – specific antibody by B lymphocytes and involve the coordinated interaction of antigen-presenting cells, T lymphocytes and B-lymphocytes. Function of T helper cells: Antigen presenting cells (APCs) present antigen on their Class II MHC molecules (MHC2). Helper T cells recognize these, with the help of their expression of CD4 co-receptor (CD4+). The activation of a resting helper T cell causes it to release cytokines and other stimulatory signals (green arrows) that stimulate the activity of macrophages, killer T cells and B cells, the latter producing antibodies. The stimulation of B cells and macrophages succeeds a proliferation of T helper cells. 3- Passive Immunity • • • • It is a form of induced immunity. Antibodies are borrowed from another sources. It usually gives short-term resistance (2 weeks – 6 months). Two types are recognized: - Natural Passive e.g. Baby in uterus (placenta) and Breast-fed babies (milk) - Artificial Passive e.g. Gamma globulin injection. Extremely fast, but short lived (e.g. snake venom) Immune system (cont’d) • • • • In some instances, the response of immune system can result in serious problems. For example, the introduction of an allograft (that is, the graft of an organ or tissue from one individual to another who is not genetically identical) can elicit a damaging immune response, causing rejection of the transplanted tissue. Transplantation of organs and tissues (for example, kidney, heart, or bone marrow) has become routine due to improved surgical techniques and better tissue typing. Another immune-related problem is the problem associated with autoimmune diseases (e.g. SLE, RA, MS, autoimmune hemolytic anemia, acute glomerulonephritis), when the immune system fail to recognize the self antigen and attack it. Immunosuppressant drug are now utilized to inhibit rejection of transplanted tissues and to treat the autoimmune diseases. Because of their severe toxicities when used as monotherapy, a combination of immunosuppressive agents (2-4 agents with different mechanisms of action), usually at lower doses, is generally employed. Immunosuppressant agents I. Selective inhibitors of cytokine production or function: 1) Calcineurin inhibitors Cyclosporine Tacrolimus (FK506) 2) Sirolimus (rapamycin). II. Inhibitors of cytokine gene expression ◦ Corticosteroids (Prednisone, Methylprednisolone, Dexamethasone) Prednisolone, III. Cytotoxic drugs • • Inhibitors of purine or pyrimidine synthesis (Antimetabolites): ◦ ◦ ◦ ◦ Azathioprine Myclophenolate Mofetil Leflunomide Methotrexate Alkylating agents: Cyclophosphamide IV. Immunosuppressive antibodies that block T cell surface molecules involved in signaling immunoglobulins ◦ antilymphocyte globulins (ALG). ◦ antithymocyte globulins (ATG). ◦ Rho (D) immunoglobulin. ◦ Basiliximab ◦ Daclizumab ◦ Muromonab-CD3 V. Interferon VI. Thalidomide I. Selective Inhibitors of Cytokine Production and Function • • • Cytokines are soluble, antigen-nonspecific, signaling proteins that bind to cell surface receptors on a variety of cells. Of particular interest when discussing immunosuppressive drugs is IL-2 cytokine, a growth factor that stimulates the proliferation of antigen-primed (helper) T cells, which subsequently produce more IL-2, IFNɣ, and TNFα. These cytokines collectively activate natural killer cells, macrophages, and cytotoxic T lymphocytes. Drugs that interfere with the production of IL-2, such as cyclosporine, and Tacrolimus (FK506) are also known as calcineurin inhibitors . Example of drugs that inhibit IL-2 action is sirolimus (rapamycin) • Cyclosporine (CsA) is a lipophillic cyclic polypeptide composed of 11 amino acids that is extracted from a soil fungus (Tolypocladium inflatum). Mechanism of action: • • • • Cyclosporine preferentially suppresses cell-mediated immune reactions, whereas humoral immunity is affected to a far lesser extent. It acts by blocking activation of T cells by inhibiting interleukin-2 production (IL-2). It also decreases proliferation and differentiation of T cells. After diffusing into the T cell, CsA binds to cytosolic protein known as cyclophilin (immunophilin) that acts as CsA intracellular receptors. Cyclosporine-immunophilin complex inhibits calcineurin, a phosphatase necessary for dephosphorylation of transcription factor NFATc (cytosolic Nuclear Factor of Activated T cells). required for interleukins synthesis (IL-2). NFATc suppresses cellmediated immunity. Pharmacokinetics: • • • Can be given orally or I.V. infusion. It is slowly and incompletely absorbed from GIT. Peak levels is reached after 2– 4 hours, elimination half life is 24 h. Oral absorption is delayed by fatty meal. CsA is usually available in soft gelatin capsules, microemulsion that has higher bioavailability-is not affected by food. • • • 50 – 60% of cyclosporine accumulates in blood (erythrocytes & lymphocytes). CsA is extensively metabolized by liver CYP-450, primarily by hepatic CYP3A4. Excreted mainly through the biliary route, and about 6% is excreted in urine. • Therapeutic Uses: • • • CsA is used to prevent rejection of kidney, liver, and cardiac allogeneic transplants. CsA is most effective in preventing acute rejection of transplanted organs when combined in a double-drug or tripledrug regimen with corticosteroids and an antimetabolite such a smycophenolate mofetil. CsA is used for the treatment autoimmune disorders like severe, active rheumatoid arthritis, and recalcitrant psoriasis that does not respond to other therapies. Adverse Effects: • • • • • • • • • • • Many of the adverse effects caused by CsA are dose dependent; therefore, it is important to monitor blood levels of the drug Nephrotoxicity; critical to monitor kidney function. Reduction of the CsA dosage can result in reversal of nephrotoxicity in most cases, although nephrotoxicity may be irreversible in 15% of patients. Symptoms of nephrotoxicity is enhanced by co-administeration with NSAIDs and aminoglycosides antibiotics. Liver dysfunction. Hypertension, hyperkalemia.(K-sparing diuretics should not be used). Hyperglycemia. Multiple infections especially viral infections (Herpes cytomegalovirus). Lymphoma (Predispose recipients to cancer). Hirsutism Neurotoxicity (tremor). Gum hyperplasia. Anaphylaxis may experienced after I.V. Drug Interactions • • Clearance of cyclosporine is enhanced by co-administration of CYP450 inducers (Phenobarbitone, Phenytoin & Rifampin rejection of transplant. Clearance of cyclosporine is decreased when it is coadministered with erythromycin or Ketoconazole, Grapefruit juice cyclosporine toxicity. • Tacrolimus (TAC, originally called FK506) is a macrolide lactone antibiotic that is isolated from a soil actinobacteria (Streptomyces tsukubaensis) Mechanism of action: • TAC exerts its immunosuppressive effect in the same mechanism as CsA, except that it binds to a different immunophilin protein , FKBP-12 (FK506-binding protein12) creating a new complex (tacrolimus- FKBP12 complex) that inhibits calcineurin. Pharmacokinetics: • • • • • • • • TAC may be administered orally , I.V., or some topical preperations (ointment). GI absorptin of TAC is incomplete and variable, reduced by fat and carbohydrate meals. TAC is from 10- to 100-fold more potent than CsA. It has half-life after I.V. adminsteration is 9-12 hours. It is highly bound to plasma proteins and is also concentrated in erythrocytes. It is undergoes hepatic metabolism by the CYP3A4 isozyme; thus, the same drug interactions with CsA occur. At least one metabolite of TAC has been shown to have immunosuppressive activity. Excreted mainly in bile and minimally in urine. Therapeutic Uses: as cyclosporine • • • Organ and stem cell transplantation Prevention of rejection of liver and kidney transplants (given with a corticosteroids and/or an antimetabolite). TCA is also used in treatment of dermal symptoms associated with some autoimmune disorders (e.g. dermatitis and psoriasis). Adverse Effects: • • • • • • • • Nephrotoxicity (more than CsA) Neurotoxicity; tremor, seizures, and hallucinations, (more than CsA) insulin-dependent diabetes mellitus GIT disturbances Hperkalemia Mild hypertension Anaphylaxis NO hirsutism or gum hyperplasia Drug interactions: as CsA What are the differences between CsA and TAC ? TAC is more favorable than CsA due to: • • • TAC is 10 – 100 times more potent than CsA in immunosupression capacity. TAC has decreased episodes of rejection. TAC is combined with lower doses of glucocorticoids., But • • • • TAC is more nephrotoxic and neurotoxic. TAC has also been found to have a lower incidence of cardiovascular toxicities such as hypertension and hyperlipidemia, both of which are common disease states found in kidney transplant recipients. Sirolimus (SRL) is macrolide antibiotic It was first discovered as a product of the bacterium (Streptomyces hygroscopicus ) in a soil sample from a pacific island known as Rapa Nui, hence it was initially marketed under the name Rapamycin Mechanism of action: • • • • Sirolimus is to bind the cytosolic protein FKBP12 (in a manner similar to tacrolimus), but instead of inhibition of calcineurin, Sirolimus-FKBP12 complex inhibits the mammalian target of rapamycin1 (mTOR). mTOR is a serine-threonine kinase that is essential for many cellular functions, such as cell-cycle progression, DNA repair, and regulation of protein translation Binding of Sirolimus-FKBP12 complex to mTOR blocks the activated T -cells proliferation, B cell proliferation & immunoglobulin production. Unlike CsA and TAC, SRL does not owe its effect to lowering IL-2 production but, rather, to inhibiting the cellular responses to IL-2. Pharmacokinetics: • The drug is available only as oral preparations. • It is readily absorbed from GIT (rate is decreased by high-fat meals) • A loading dose is required at the time of initiation of therapy. • SRL is extensively bound to plasma proteins (~ 92%) and has a long half-life (57-63 hours) compared to those of CsA and TAC. • SRL is metabolized by the CYP3A4 isozyme and has the same drug-drug interactions as CsA and TAC. • Due to competition in protein binding sites, SRL increases the drug concentrations of CsA, and careful blood level monitoring of both agents must be employed to avoid harmful drug toxicities uppon combined therapy. • SRL and its metabolites are predominantly eliminated in the feces. Therapeutic Uses: SRL has both anti-proliferative and immunosuppressive effects (equipotent to CsA). • • • • • In renal transplantation(used mainly combined with CsA and corticosteroids) To limit the long-term side effects of the calcineurin inhibitor, SRL is often utilized in calcineurin inhibitor withdrawal protocols in patients who remain rejection free during the first 3 months posttransplant. Heart allografts In halting graft vascular disease (SRL-coated stents inserted into the cardiac vasculature inhibit restenosis of the blood vessels by reducing proliferation of the endothelial cells). Hematopoietic stem cell transplant recipients. Adverse Effects: • • • • • • Hyperlipidemia (elevated cholesterol and triglycerides), The combination of CsA and SRL or TAC and SRL are more nephrotoxic than CsA or TAC alone headache, nausea thrombocytopenia. and diarrhea, leukopenia, and Impaired wound healing especially in obese and diabetic patients which may represents a problem immediately following the transplant surgery. The corticosteroids were the first pharmacologic agents to be used as immunosuppressives both in transplantation and in various autoimmune disorders. They have both anti-inflammatory immunosuppressant effects. action and II. Inhibitors of Cytokine gene expression • • • • • The corticosteroids were the first pharmacologic agents to be used as immunosuppressives both in transplantation and in various autoimmune disorders. They have both anti-inflammatory immunosuppressant effects. action and They are still one of the mainstays for attenuating rejection episodes. The most common agents are Prednisone, prednisolone, methylprednisolone, and dexamethasone Mechanism of action: On entering cells, they bind to the glucocorticoid receptor (GR). The glucocorticoid-GR complex translocates into the nucleus and interacts with DNA to regulates the gene transcription of . Among the genes affected are those involved in inflammatory responses. - Decrease production of inflammatory mediators as prostaglandins, leukotrienes, histamine, PAF, bradykinin mainly by their inhibitory action on annexin-1 (a phoshplipase A inhibitor). - Decrease production of a wide array of proinflammatory cytokines like IL-1, IL-2,IL-3,…, TNF-, GM-CSF. - Stabilize lysosomal membranes. - Decrease generation of IgG, and nitric oxide. - Inhibit antigen processing by macrophages. - Suppress T-cell helper function - Decrease T-lymphocyte proliferation. Pharmacokinetics: - Corticosteroids may be administered by a variety of routes. Most are active when given orally, and parentrally . In addition topical preparations, eye/ear/ nasl drops and aerosoles are also available. - Corticosteroids (~90%) bind to corticosteroid-binding globulin (CBG) and to albumin with variable tendencies. - Plasma half-life 2-8 hours, with variable durations depending on preparation ;(8-12 hrs for SAGCs e.g. Cortisol, 18-36 hrs for IAGCs e.g. Predinsolne, and 36-54 hrs for LAGCs e.g. Dexamethasone, Betamethasone). - Generally GCs are metabolized in the liver by both microsomal (mainly CYP3A4 & CYP2D6) and nonmicrosomal enzymes, where they are reduced and conjugated (glucourinate and sulfate), forming inactive water-soluble metabolites that are excreted by the kidney (~75%) in addition to ~25% are execreted in feces. - Some GCs are bioactivated after adminstration like Prednisone that is converted in vivo to active metabolite prednisolone. Therapeutic Uses: • • • Corticosteroids are first line therapy for solid organ allografts & haematopoietic stem cell transplantation to attenuating rejection episodes. Autoimmune diseases as refractory rheumatoid arthritis, systemic lupus erythematosus, temporal arthritis, and asthma. Acute or chronic rejection of solid organ allografts. Adverse Effects: • The use of these agents is associated with numerous adverse effects. For example, ◦ Hypercholesterolemia ◦ Hyperglycemia ◦ Hypertension ◦ Cataract ◦ Osteoporosis ◦ Increase liability to infection ◦ Adrenal suppression III. Cytotoxic agents • Alkylating agents e.g. Cyclophosphamide • Inhibitors of purine or pyrimidine synthesis (Antimetabolites): Azathioprine Myclophenolate Mofetil Leflunomide Methotrexate - • • Cyclophosphamide is a cytotoxic agent acts by alkaylation of the DNA. Cyclophosphamide is a prodrug that is undergo some modification by the effect of liver CYP450 to form the active compounds, phosphoramide mustard and acrolein. Reaction of the phosphoramide mustard with DNA is considered to be the cytotoxic step especialy to those rapidly deviding cells like neoplastic cells,proliferating lymphoid cells and germ cells. Pharmacokinetics: • • It is administered orally& intravenously Cyclophosphamide is converted by mixed-function oxidase enzymes (cytochrome P450 system; see chart) in the liver to active metabolites.The main active metabolite is 4hydroxycyclophosphamide, and aldophosphamide. Most of the aldophosphamide is oxidised by the enzyme aldehyde dehydrogenase (ALDH) to make carboxyphosphamide. A small proportion of aldophosphamide freely diffuses into cells, it is decomposed into two compounds, phosphoramide mustard and acrolein. • The execretion occurs equally between renal and bilary routs. Therapeutic Uses: • • It is widely utilized in treatment of a wide variety of neoplastic diseases (anticancer), such aslymphoma and breast cancer. Immunosupressant in autoimmune disorders such as rheumatoid arthritis , systemic lupus erythrematosus, Autoimmune hemolytic anemia Adverse effects: • • • Alopecia and GIT disorders (Nausea -vomiting-diarrhea) Bone marrow suppression Hemorraghic cystitis , which can lead to fibrosis of the bladder. The latter toxicity has been attributed to acrolein which is toxic to the bladder epithelium. This can be prevented through the use of aggressive hydration and/or MESNA (sodium 2mercaptoethane sulfonate), which neutralizes the toxic metabolites, minimizes this problem. • • Sterility (testicular atrophy and aspermia in male& amenorrhea in females) Veno-occlusive disease of the liver Antimetabolites 1- Azathioprine It is a purine analogue immunosuppressive drug that was the first agent to achieve widespread use in organ transplantation. • • • • It is a prodrug that is converted first to 6-mercaptopurine (6MP) and then to the corresponding nucleotide, thioinosinic acid. Mechanism of action: The immunosuppressive effects of azathioprine are due to this nucleotide analog. Because of their rapid proliferation in the immune response, lymphocytes (B &T) are predominantly affected by the cytotoxic effects of azathioprine. Azathioprine Inhibits de novo synthesis of purines required for lymphocytes proliferation. Pharmacokinetics: • • • • It is administered orally or intravenously. Widely distributed but does not cross BBB. Metabolized in the liver to 6-mercaptopurine or to thiouric acid (inactive metabolite) by xanthine oxidase. Excreted primarily in urine. Therapeutic Uses: • • It is used to prevent organ rejection following organ transplantation To treat a vast array of autoimmune diseases, including rheumatoid arthritis, Systemic lupus erythematosus inflammatory bowel disease (such as Crohn's disease and ulcerative Colitis), multiple scleross, autoimmune hepatitis, atopic dermatitis, Myasthenia Gravis, Acute glomerulonephritis, and others. Adverse effects: • • • • Bone marrow suppression (leukopenia&thrombocytopenia). Nausea , vomiting and diarrhea Hepatotoxicity. Increased risk of infections and mutagencity (Lymphoma) Drug Interactions: • • Concomitant use with ACE inhibitors in renal transplant patients can lead to an exaggerated leukopenic response. Allopurinol, an inhibitor of xanthine oxidase that is used to treat gout, significantly inhibits the metabolism of azatihioprine; therefore, the dose of azathioprine must be reduced by 60 to 75 percent. 2- Mycophenolates • Both Mycophenolate mofetil (MMF) and mycophenolate sodium (MPS) are semisynthetic derivative of mycophenolic acid from fungus source. • Both are Prodrugs that are rapidly hydrolyzed in the gastrointestinal tract to mycophenolic acid (MPA) Mechanism of action: • • Inhibit de novo synthesis of purines (especially guanosine nucleotides). MPA is a potent inhibitor of inosine monophosphate dehydrogenase (IMPDH), crucial for purine synthesis deprivation of proliferating T and B cells of nucleic acids. Pharmacokinetics: • • • • • • It is given orally, I.V or I.M. Dose 2-3 g/day. Rapidly and completely absorbed after oral administration. It undergoes first-pass metabolism to give the active moiety, mycophenolic acid (MPA). MPA is extensively bound to plasma protein. metabolized in the liver by glucuronidation. Excreted in urine as glucuronide conjugate Therapeutic uses: • • • • Solid organ transplants (heart, kidney, and liver transplants). Steroid-refractory hematopoietic stem cell transplant patients. Combined with prednisone as alternative to CSA or tacrolimus. Rheumatoid arthritis & dermatologic disorders. Adverse effects & drug interaction: • • • • • GIT toxicity: Nausea, Vomiting, diarrhea, abdominal pain. Leukopenia, neutropenia & anemea Higher risk of CMV infection. It is CI to be administered during pregnancy Concomitant administration with antacids containing Mg2+ or Al 3+, or with cholestyramine, can decrease absorption of the drug. 3- Leflunamide • • • Leflunomide is a synthetic disease-modifying antirheumatic group of drugs- antimetabolite immunosupressant (DMARD). It acts by inhibiting dihydroorotate dehydrogenase (an enzyme involved in de novo pyrimidine synthesis) It is a prodrug ,following oral administration, leflunomide is metabolized to teriflunomide, which is responsible for all of the drug's activity in vivo. Pharmacokinetics: • • • The drug is given orally. It is rapidly metabolized to its pharmacologically- active metabolite teriflunomide. Teriflunomide is metabolized in the liver and excreted as well renally and billary. After oral administration, peak plasma levels of teriflunomide occurred between 6 and 12 hours after dosing. Due to its very long half-life (approximately 2 weeks), a loading dose of 100 mg for 3 days is required to reach steady-state levels quickly. - Therapeutic uses: Leflunomide has both immunosuppressant and antiinflammatory effects. Leflunomide has been approved for the treatment of rheumatoid arthritis or psoriatic arthritis. It slows the progression of the disease and relief the associated symptoms such as joint tenderness and decreased joint and general mobility in human patients. Adverse effects: • • • • • • • Elevation of liver enzymes Renal impairment Teratogenicity (Women or men should not have babies before 2 years after termination of therapy have elapsed or undergo a rapid wash-out procedure like an eleven-day scheme with oral cholestyramine or the use of activated charcoal is indicated and will soon decrease plasma levels below the critical limit of 0.02 mg/l ). Cardiovascular effects (tachycardia). Leflunomide has the potential to myeloid/lymphatic malignancies or solid cancers promote Methotrexate (MTX) is synthetic antimetabolite used mainly in treatment of cancer, and autoimmune diseases It acts by inhibiting the metabolism of folic acid. It Inhibits DHFR enzyme required for folic acid activation (tetrahydrofolic) and as a result Inhibition of DNA, RNA &protein synthesis occur. Pharmacokinetics: • • It can be taken orally or administered by injection (I.M., I.V., S.C., or I.T.). Methotrexate is metabolized by intestinal bacteria to the inactive metabolite 4-amino-4-deoxy-N-methylpteroic acid (DAMPA), which accounts for less than 5% loss of the oral dose Oral doses are taken weekly. It is excreted in urine. Therapeutic uses: • • • Cancer Chemotherapy: In the treatment of a number of cancers including: breast, head and neck, leukemia, lymphoma, lung, osteosarcoma, and bladder. Autoimmune disorders: It is used as a treatment for some autoimmune diseases including: psoriasis and psoriatic arthritis, Crohn's disease, and rheumatoid arthritis. Pregnancy termination: Methotrexate is commonly used (generally in combination with misoprostol) to terminate pregnancies during the early stages (i.e., as an abortifacient). It is also used to treat ectopic pregnancies. Adverse effects & drug interaction: • • Nausea, vomiting, stomatitis. diarrhea, Fatigue, fever, and dizziness abdominal pain, ulcerative • • • • • • Alopecia Bone marrow depression (low white blood cell count) Predisposition to multiple infection Pulmonary fibrosis Renal & hepatic disorders Methotrexate is a highly teratogenic drug and contrindicated during pregnancy. IV. Immunosuppressive antibodies - The use of antibodies plays a central role in prolonging allograft survival. Most of them block T cell surface molecules involved in signaling immunoglobulins. They include: - Antithymocyte globulins (ATG). - Muromonab-CD3 - Rho (D) immunoglobulin. - Basiliximab - Daclizumab - Infliximab Preparation: 1. By immunization of either rabbits or horses with human lymphoid cells producing a mixture of polyclonal antibodies directed against a number of lymphocyte antigens (variable, less specific). 2. Hybridoma technology: produce antigen-specific, monoclonal antibody (homogenous, specific). • Hybridomas are produced by fusing mouse antibodyproducing cells with human immortal, malignant plasma cells. Hybrid cells are selected and cloned, and the antibody specificity of the clones is determined. Clones of interest can be cultured in large quantities to produce clinically useful amounts of the desired antibody. 3. Recombinant DNA technology: • • Antibodies can be obtained by replace part of the mouse gene sequence with human genetic material thus humanized antibody produced (less antigenicity-longer half life). The names of monoclonal antibodies conventionally contain “Muro-” in their names are from a murine (mouse) source or contain “zu” or “xi” if they are humanized . 1-Antithymocyte globulins (ATG) • • • • Thymocytes are cells that develop in the thymus and serve as T-cell precursors. The antibodies developed against them are prepared by immunization of large rabbits or horses with human lymphocytes (Polyclonal antibodies). The antibodies bind to the surface of circulating T lymphocytes, which then undergo various reactions, such as antibody-dependent cytotoxicity, and apoptosis. The antibody-bound cells are phagocytosed in the liver and spleen, resulting in lymphopenia and impaired T-cell responses & cell-mediated immunity. Pharmacokinetics: • • The antibodies are slowly infused I.V. Their half-life extends from 3 to 9 days. Therapeutic uses: • They are primarily employed, together with other immunosuppressive agents (like cyclosporine), at the time of transplantation to prevent early or acute allograft rejection, or they may be used to treat severe rejection episodes . • • Corticosteroid-resistant Acute allograf rejection. Adverse effects: • • • Hypersenstivity (chills and fever, skin rashes) leukopenia and thrombocytopenia, Tendency to infections especially CMV or other viruses. 2- Muromonab-CD3 • • • Muromonab-CD3 is a murine monoclonal antibody that is synthesized by hybridoma technology and directed against cell surface glycoprotein CD3 antigen of human T cells. Binding to CD3 (antigen recognition site) leading to disruption of T-lymphocyte function, and depletion of. circulating T cells with decreased immune response. Because muromonab-CD3 recognizes only one antigenic site, the immunosuppression is less broad than that seen with the polyclonal antibodies. T cells usually return to normal within 48 hours of discontinuation of therapy. Pharmacokinetics: • • The antibody is administered I.V., initial binding of muromonab-CD3 to the antigen transiently activates the T cell and results in cytokine release (cytokine storm). It is therefore usual to premedicate the patient with Prednisolone, diphenhydramine , and acetaminophen to alleviate the cytokine release syndrome. • The antibody is metabolized and excreted in the bile. Therapeutic uses: • • • Muromonab-CD3 is used for treatment of acute rejection of renal allografts For treatment of corticosteroid-resistant allograft rejection in cardiac and hepatic transplant patients. It is also used to deplete T cells from donor bone marrow prior to transplantation. 3- Rho (D) immune globulin • • Rho (D) is a concentrated solution of human IgG containing higher titer of antibodies against Rho (D) antigen of red cells. In a Rh negative mother Rho(D) Immune Globulin can prevent temporary sensitization of the maternal immune system to Rh D antigens, which can cause rhesus disease in the current or in subsequent pregnancies. Given to Rh-negative mother within 24-72 hours after delivery or miscarriage of Rh-positive baby (2 ml, I.M.) to prevent development of an immunological condition known as Rhesus disease (or hemolytic disease of newborn; erythroblastosis fetalis) of the next Rh positive babies. Adverse Effects: - Local pain - Fever 4- Alemtuzumab • • • • A humanized monoclonal antibody directed against surface protein (CD52) – expressed on lymphocytes, monocytes, macrophages, natural killer cells and thymocytes It exerts its effects by causing profound depletion of T cells from the peripheral circulation as well as peripheral lymph nodes. it is being utilized in combination with low-dose CNIs or sirolimus especially, in corticosteroid avoidance protocols in renal and liver transplantation especially in HCV-infected subjects. Adverse effects: include Cytokine storm syndrome, requiring premedication with acetaminophen, and diphenhydramine, in addition to neutropenia, and anemia. 5- IL-2-receptor antagonists • • • • The antigenicity and short serum half-life of the murine monoclonal antibody have been averted by replacing most of the murine amino acid sequences with human ones by genetic engineering. Basiliximab is a chimeric human-mouse monoclonal IgG (25% murine, 75% human protein). Daclizumab is a humanized monoclonal IgG (90% human protein). They have less antigenicity & longer half lives than murine antibodies Mechanism of action: • • • Both compounds are anti-CD25 antibodies and bind to CD25 (α-subunit chain of IL-2 receptor on activated lymphocytes) They thus block IL-2 stimulated T cells proliferation and & Tcell response to any antigenic stimulus interfere with the proliferation of these cells. Basiliximab is more potent than daclizumab as a blocker of IL-2 stimulated T-cell replication. Therapeutic uses: • • Both agents have been used for prophylaxis of acute rejection in renal transplantation in combination with CsA and corticosteroids. They are not used for the treatment of ongoing rejection. Pharmacokinetics: • • Both antibodies are given intravenously. The serum half-life of daclizumab is about 20 days, Five doses of daclizumab are usually administered (the first at 24 hours before transplantation, and the next four doses at 14-day intervals). • The serum half-life of basiliximab is about 7 days. Usually, two doses of this drug are administered (the first at 2 hours prior to transplantation, and the second at 4 days after the surgery). Adverse effects: Both Daclizumab and Basiliximab are well tolerated. Their major toxicity is GI upset, mild hypersensitivity symptoms with minimal risk of opportunistic infections and post-transplant lymphoproliferative disorder. 6- Anti TNF-α therapy • • • TNF-α receptor antibodies approved for clinical use are infliximab , certolizumab,and adalimumab chimeric humanmouse monoclonal IgGs that targets variable components of TNF- α receptors (both soluble and cell-membraneIt bound) and neutralize them and inhibits further activity. Etanercept is a humanized fusion protein linking mimics inhibitory effects of naturally occurring soluble TNF receptors. Mainly used in treatment of autoimmune disorders. Adverse effects: - Infusion reaction – fever, urticaria, hypotension, dyspnoea - Opportunistic infections – TB, RTI, UTI V- Interferones • • • • • Interferons are a family of naturally occurring, inducible glycoproteins cytokines that are used clincally in many purposes. They have different immunomodulating actions. The interferons are synthesized by recombinant DNA technology. Three types of interferon exist, α , β, and γ. IFN- γ : Acts as immunostimulant. It increases expression of MHC molecules, enhances the activity of macrophages and natural killer cell, and stimulates production of IgG. IFN- α, β : Act as inhibitors of cellular proliferation (cancer cells, Immune cells and viral infections) Therapeutic uses: - Treatment of certain infections e.g. Hepatitis B&C (IFN- α ). - Certain forms of cancer e.g. melanoma, renal cell carcinoma (IFN- α ). - Autoimmune diseases e.g. Rheumatoid arthritis (IFN- α & β ). - Multiple sclerosis (IFN- β): reduced rate of exacerbation. Adverse effects: Fever, chills, myelosuppression. Immunosstimulants Immunosstimulants are substances that stimulate the immune system by inducing activation or enhancing the activity of any of its components. Immunosstimulant agents are now utilized to enhancing cellular and/or humoral immunity should benefit people with immune deficiencies (primary and secondary) and severe infections and cancers. Degree of stimulation relatively small Types of immunostimulant agents: 1- Synthetic drugs: e.g. Levamisole and Thalidomide 2- Immunostimulant vaccines: e.g. BCG vaccine 3- Recombinant cytokines : e.g. IFN-,GM-CSF, IL-2 4- Immunization vaccines (active and passive) 5-Immunostimulatory MAbs I-Immunostimulant drugs 1- Levamisole Levamisole (LMS) is an imidazothiazole derivative that has an immunomostimulator and anthelminthic activities .It is usually used as phoshates or hydrochloride salts. It has been used in humans to treat many parasitic worm infections, In addition to its utilization as immunostimulant in combination with some chemotherapeutic agents (like 5-FU) to treat many types of human cancers like colon cancer, melanoma, and head and neck cancer. Levamisole’s mechanism of action for its immunostimulating effects are not well understood. It is believed it enhances cell-mediated immune function in peripheral T- and B-lymphocytes and stimulates phagocytosis by monocytes. Its immune stimulating effects appear to be more pronounced in the immune-compromised individuals. Levamisole may also interferes with the growth of cancer cells and slows their growth and spread in the body by blocking carbohydrate metabolism. Side effects include GI upset, loss of appetite, change in taste and smell, muscle aches, fatigue, dizziness, headache and skin rash. Alcohol and alcohol containing products should be avoided while taking levamisole. Flushing, nausea, vomiting, stomach pain headache, swelling, and rashes can occur. 2-Thalidomide Thalidomide was first introduced as a sedative and antiemetic drug during the period of world war II, it was withdrawn due to sever side effects “teratogenicity and neuropathy”. Now there are revived interests in using thalidomide as immunostimulating agent used to treat a number of medical conditions like cancer (e.g. multiple myeloma) and tough infectious diseases like leprosy. Thalidomide has many mechanisms by which it exerts the immunostimulatory effect: - It stimulates T-cells proliferation - It enhances activation of NK cells - It stimulate production of IL-2& IFN-γ and inhibits the production of other cytokines (e.g. IL-10 ). - Thalidomide has antiangiogenic effect (inhibits the growth of new blood vessels), which makes it be useful in treating some types of cancer like multiple myloma, prostate cancer and lymphomas. Thalidomide is contraindicated in women within childbearing potential due to the high risk of teratogenicity. II-Bacterial-derived Immunostimulant Vaccines Bacillus Calmette-Guérin (BCG) vaccine BCG is a vaccine prepared from life-attenuated bovine tuberculosis bacillus, Mycobacterium bovis, that has lost its virulence in humans, but still retained its strong antigenicity enough to stimulate immune system to produce the corresponding antibodies. BCG is used predominantly as upperarm I.D. injection for vaccination against tuberculosis. BCG is also used as intravesical therapy in the treatment of superficial forms of bladder cancer and colorectal cancer. The mechanism is unclear, it appears a local immune reaction is mounted against the tumor (may act in part by stimulating TNF-α release from macrophages). A number of cancer vaccines in development use BCG as an adjuvant to provide an initial stimulation of the patients' immune system. The most noticeable adverse effects associated with BCG are causes some pain and scarring at the site of injection, hypersensitivity, chills, fever, skin rashes, hypotension and may be shocking. III- Cytokines • Human cytokines prepared by recombinant DNA technology 1- human recombinant IL-2 (rhIL-2 ); aldesleukin, proleukin It binds to the IL-2 receptor on T-lymphocytes It stimulates the proliferation and diffrentiation of antigen-primed (helper) T cells, which subsequently produce more cytokines( IL-2,IL1, IFNɣ, and TNFα). These cytokines collectively activate natural killer cells, macrophages, and cytotoxic T lymphocytes. rhIL-2 is used in metastatic renal cell cancer and melanoma. Adverse effects: Inflammation and increased vascular permeability, peripheral edema, fever and hypotension. Cardiac arrhythmia, anemia, nausea, vomiting, diarrhea, confusion 2- Interferone (INF-ɣ) INF-ɣ boosts the activity of macrophages and NK cells. It stimulates the expression of MHC molecules and production of IgG. It causes cancer cells to produce more antigens , suppresses the growth of cancer cells and inhibits angiogenesis It is approved for use with some leukemias, lymphoma, renal cancer and melanoma in addition to prevention of secondary infections in chronic granulomatous diseases. Adverse effects: Flu-like symptoms – fever, chills, headache, GI disturbance Hypotension, Arrhythmia CNS- depression and confusion 3-Granulocyte-macrophage colony-stimulating factor (GM-CSF) GM-CSF is a cytokine secreted mainly by macrophages, mast cell in addition to endothelial cells, and fibroblasts. It functions as a white blood cell growth factor. rhGM-CSF interacts with specific cell-surface receptors found on various immune component cells. GM-CSF stimulates stem cells to produce granulocytes (neutrophils, eosinophils, and basophils) and monocytes. It also activates the phagocytic activity of mature monocytes and prolongs their survival in the circulation. GM-CSF is used in cancer patients after chemotherapy to treat neutropenia and increased risk of serious infection receiving cancer chemotherapy It is also used to used to accelerate myeloid recovery after autologous BMT in patients with lymphoma, and acute lymphoblastic leukemia Adverse effects: include fever, malaise, arthralgias, myalgias, and a capillary leak syndrome characterized by peripheral edema and pleural or pericardial effusions. Allergic reactions may occur but are infrequent. Spleenomegaly is a rare but serious complication of the use of GM-CSF. IV- Immunization vaccines • • • • • Vaccines are biological preparations that improves immunity. It could act against particular disease or boost the general immune function. Active immunization (vaccination): include administration of the antigen of the disease-causing microorganism in the form of weakened or killed microbe , its toxins or a specific protein or peptide constituent of an organism that stimulate the immune system to recognize these antigens and generate an adaptive immune response by raising the specific antibodies to destroy it, and "remember" it, so that the immune system can more easily recognize and destroy any of these microorganisms that it later encounters. the response takes days/weeks to develop but may be long lasting even lifelong. Vaccines can be prophylactic to prevent or ameliorate the effects of a future infection by pathogen (e.g. polio vaccine, MMR vaccine)., or therapeutic (e.g. vaccines against cancer) Passive immunization: fortification of the immune system by administration of prepared imunglobulines antibodies to protect against infection; it gives immediate, but short-lived protection for several weeks to 3 or 4 months at most. It is mainly used as replacement therapy in individuals with primary immune deficiency diseases . Also, to manage some acute infections when active vaccination is inadequate Passive immunity is usually classified as natural or acquired. The transfer of maternal tetanus antibody (mainly IgG) across the placenta provides natural passive immunity for the newborn baby for several weeks/months until such antibody is degraded and lost. In contrast, acquired passive immunity refers tothe process of obtaining serum from immune individuals, pooling this, concentrating the immunoglobulin fraction and then injecting it to protect a susceptible person. • Examples: vaccines of hepatitis B, botulism, diptheria, tetanus, rabies V- Immunostimulatory MAbs Monoclonal antibodies prepared against surface antigenic marker proteins specific for tumor cells (e.g. anti-HER2/Neu, anti-VEGF) mAbs act directely when binding to a cancer specific antigens and induce immunological response to cancer cells. Such as inducing cancer cell apoptosis, inhibiting growth, or interfering with a key function. mAbs also was modified to deliver a toxin, radioisotope, cytokine or other active conjugates into cancer cells.