final immunomodulators & immunosuppressants

Upload: sumanth-kumar-reddy

Post on 30-Oct-2015

73 views

Category:

Documents


0 download

TRANSCRIPT

  • 1

    Sumanth Dept of Pharmacology

    Immunosuppressants & Immunomodulators

    Abnormal Immune Responses

    Whereas the normally functioning immune response can successfully neutralize

    toxins, inactivate viruses, destroy transformed cells, and eliminate pathogens.

    Inappropriate responses can lead to extensive tissue damage (Hypersensitivity)

    or reactivity against self-antigens (Autoimmunity); conversely, impaired

    reactivity to appropriate targets (Immunodeficiency) may occur and abrogate

    essential defense mechanisms.

    Autoimmunity

    Autoimmune disease arises when the body mounts an immune response against

    itself due to failure to distinguish self-tissues and cells from foreign (nonself)

    antigens or loss of tolerance to self.

    This phenomenon derives from the activation of self-reactive T and B

    lymphocytes that generate cell-mediated or humoral immune responses directed

    against self-antigens.

    The pathologic consequences of this reactivity constitute several types of

    autoimmune diseases.

    Autoimmune diseases are highly complex due to MHC genetics, environmental

    conditions, infectious entities, and dysfunctional immune regulation.

    Examples of such diseases include

    Rheumatoid arthritis

    Systemic lupus erythematosus

    Multiple sclerosis

    Insulin-dependent diabetes mellitus (type 1 diabetes).

    In Rheumatoid arthritis, IgM antibodies (rheumatoid factors) are produced that

    react with the Fc portion of IgG and may form immune complexes that activate

    the complement cascade, causing chronic inflammation of the joints and kidneys.

    In systemic lupus erythematosus, antibodies are made against DNA, histones, red

    blood cells, platelets, and other cellular components.

    In multiple sclerosis and type 1 diabetes, cell-mediated autoimmune attack

    destroys myelin surrounding nerve cells and insulin-producing islet beta cells of

    the pancreas, respectively.

    In type 1 diabetes, activated CD4 TDTH cells that infiltrate the islets of Langerhans

    and recognize self islet beta cell peptides are thought to produce cytokines that

    stimulate macrophages to produce lytic enzymes, which destroy islet beta cells.

    Autoantibodies directed against the islet beta cell antigens are produced, but do

    not contribute significantly to disease.

  • 2

    Sumanth Dept of Pharmacology

    A number of mechanisms have been proposed to explain auto-immunity:

    Exposure of antigens previously sequestered from the immune system (eg, lens

    protein, myelin basic protein) to self-reactive T lymphocytes.

    Molecular mimicry by invading pathogens, in which immune responses are

    directed at antigenic determinants on pathogens that share identical or similar

    epitopes with normal host tissue.

    This phenomenon occurs in rheumatic fever following Streptococcus pyogenes

    infection, in which heart damage is thought to arise from an immune response

    directed against streptococcal antigens shared with heart muscle.

    Inappropriate expression of class II MHC molecules on the membranes of cells

    that normally do not express class II MHC (eg, islet beta cells).

    Increased expression of MHC II may increase presentation of self-peptides to T

    helper cells, which in turn induce CTL, TDTH, and B-lymphocyte cells that react

    against self-antigens.

    Examples of Autoimmune Diseases, Categorised by Type of Tissue Damage

  • 3

    Sumanth Dept of Pharmacology

    Pathophysiology

    Transplantation

    Currently, most organ transplantation occurs between unrelated individuals.

    Donor and recipient tissues express different MHC class I molecules, and recipient

    immune cells therefore recognize the transplanted tissues as foreign.

    This is termed Alloimmunity, and it occurs when the recipients immune system

    attacks a transplanted organ.

    In the case of a bone marrow or stem cell transplant, Graft-versus-host disease

    (GVHD) can result when donor lymphocytes mount an assault on recipient

    tissues.

    Solid Organ Rejection

    Transplant rejection of solid organs can be divided into three major phases

    according to the time to onset:

    Hyperacute rejection

    Acute rejection

    Chronic rejection

    are caused by different mechanisms and are therefore treated differently.

    Hyperacute Rejection

    Hyperacute rejection is mediated by preformed recipient antibodies against donor

    antigen. Because these antibodies are present at the time of organ implantation,

    hyperacute rejection occurs almost immediately after reperfusion of the

    transplanted organ.

    In fact, the surgeon can observe the changes in the organ minutes after restoration

    of blood flow.

    The normal, healthy, pink appearance of the transplanted organ rapidly becomes

    cyanotic, mottled, and flaccid.

    This rapid change is the result of complement activation by antibody binding to

    endothelial cells of the transplanted organ, resulting in thrombosis and ischemia.

    Most commonly hyperacute rejection is mediated by recipient antibodies that

    react with blood group antigens in donor organs (e.g., type AB donor in a type O

    recipient).

    Matching of blood types between donor and recipient prevent hyperacute

    rejection; therefore, drug therapy for hyperacute rejection is typically not

    necessary.

    Hyperacute rejection also occurs in xenotransplantation (i.e., organ

    transplantation between species, such as pig heart transplanted into a human

    recipient), due to the presence of preformed human antibodies that react against

    antigenic proteins and carbohydrates expressed by the donor species.

  • 4

    Sumanth Dept of Pharmacology

    Acute Rejection

    Acute rejection has cellular and humoral components.

    Acute cellular rejection is mediated by cytotoxic T Cells and causes interstitial as

    well as vascular damage.

    This cellular response is most commonly seen in the initial months after

    transplantation.

    Immunosuppression of T cells is highly effective at preventing or limiting

    activation of the recipient immune system by the transplanted organ, thereby

    preventing acute cellular rejection.

    In Acute humoral rejection, recipient B cells become sensitized to donor antigens

    in the transplanted organ and produce antibodies against these alloantigens after

    a period of 7-10 days.

    The antibody response is typically directed against endothelial cells and is thus

    also known as acute vascular rejection.

    Like acute cellular rejection, acute humoral rejection can usually be prevented by

    immunosuppression of the recipient after transplantation.

    Even with immunosuppression, however, episodes of acute rejection can occur

    months or even years after transplantation.

    Chronic Rejection

    Chronic rejection is believed to be both humoral and cellular in nature and does

    not occur until months or years after transplantation.

    Because hyperacute and acute rejection are generally well controlled by

    donor/recipient matching and immunosuppressive therapy, chronic rejection is

    now the most common life-threatening pathology associated with organ

    transplantation.

    Chronic rejection is thought to result from chronic inflammation caused by the

    response of activated T cells to donor antigen.

    Activated T cells release cytokines that recruit macrophages into the graft.

    The macrophages induce chronic inflammation that leads to intimal proliferation

    of the vasculature and scarring of the graft tissue.

    The chronic changes eventually lead to irreversible organ failure.

    Other contributing nonimmune factors can include ischemia-reperfusion injury

    and infection.

    No effective treatment regimens are currently available to eliminate chronic

    rejection.

    It is believed, however, that several experimental therapies have a reasonable

    chance of reducing chronic rejection.

  • 5

    Sumanth Dept of Pharmacology

    Graft-Versus-Host Disease (GVHD)

    Leukemia, primary immunodeficiency, and other conditions can be treated with

    bone marrow or peripheral stem cell transplantation.

    In this procedure, hematopoietic and immune function is restored after the

    patients bone marrow has been eradicated by aggressive chemotherapy and/or

    radiation therapy.

    GVHD is a major complication of allogeneic bone marrow or stem cell

    transplantation.

    GVHD is an alloimmune inflammatory reaction that occurs when transplanted

    immune cells attack the cells of the recipient.

    The severity of GVHD ranges from mild to life-threatening and typically involves

    the skin (rash), gastrointestinal tract (diarrhea), lungs (pneumonitis), and liver

    (veno-occlusive disease).

    GVHD can often be ameliorated by removing T cells from the donor bone marrow

    before transplantation.

    Mild-to-moderate GVHD can also be beneficial when donor immune cells attack

    recipient tumor cells that have survived the aggressive chemotherapy and

    radiation therapy. (In the case of leukemia, this is called the graft-versus-leukemia

    effect, or GVL.)

    Therefore, although removing donor T cells from the graft reduces the risk of

    GVHD, this may not be the best approach for marrow transplants used in

    antineoplastic therapy.

  • 6

    Sumanth Dept of Pharmacology

    Immunosuppressants

    Pharmacologic suppression of the immune system utilizes eight mechanistic approaches

    Inhibition of gene expression to modulate inflammatory responses

    Depletion of expanding lymphocyte populations with cytotoxic agents

    Inhibition of lymphocyte signaling to block activation and expansion of

    lymphocytes

    Neutralization of cytokines and cytokine receptors essential for mediating the

    Immune response

    Depletion of specific immune cells, usually via cell-specific antibodies

    Blockade of costimulation to induce anergy

    Blockade of cell adhesion to prevent migration and homing of inflammatory cells

    Inhibition of innate immunity, including complement activation

  • 7

    Sumanth Dept of Pharmacology

    1. Inhibition of gene expression

    2. Selective attack on clonally expanding lymphocyte populations

    3. Inhibition of intracellular signaling

    4. Neutralisation of cytokines & cytokine receptors required for T-cell stimulation

    5. Selective depletion T cells

    6. Inhibition of costimulation by antigen-presenting cells

    7. Inhibition of lymphocyte-target cell interactions

    Classification

    Inhibitors of Gene Expression: Glucocorticoids

    Cytotoxic Agents:

    Antimetabolites: Azathioprine (AZA), Methotrexate (MTX),

    Mycophenolic Acid & Mycophenolate Mofetil (MPA), Leflunomide

    Alkylating Agents: Cyclophosphamide

    Specific Lymphocyte-Signaling Inhibitors:

    Cyclosporine

    Tacrolimus

    mTOR Inhibitors: Sirolimus, Everolimus, Zotarolimus

    Cytokine Inhibition:

    TNF- Inhibitors: Etanercept, Infliximab, Certolizumab pegol,

    Adalizumab, Golimumab

    IL-12/IL-23p40 Inhibitors: Ustekinumab

    IL-1 Inhibitors: Anakinra, Rilonacept, Canakinumab

    Cytokine Receptor Antagonists: Tocilizumab

    Depletion of Specific Immune Cells:

    Polyclonal Antibodies: Antithymocyte globulin (ATG)

    Monoclonal Antibodies

    Anti-CD3: OKT3 (Muromonab-CD3)

    Anti-CD20 mAb: Rituximab

    Anti-CD25 mAb: Daclizumab, Basiliximab

    Anti-CD52 mAb: Alemtuzumab

    LFA-3: LFA-3(CD58), Alefacept

    Inhibition of Costimulation: Abatacept, Belatacept

    Blockade of Cell Adhesion: Natalizumab

    Inhibition of Complement Activation: Eculizumab

  • 8

    Sumanth Dept of Pharmacology

    Glucocorticoids

    Glucocorticoids (corticosteroids) were the first hormonal agents recognized as

    having lympholytic properties.

    Administration of any glucocorticoid reduces the size and lymphoid content of

    the lymph nodes and spleen, although it has no toxic effect on proliferating

    myeloid or erythroid stem cells in the bone marrow.

    Prednisone, Prednisolone, and other glucocorticoids are used alone and in

    combination with other immunosuppressive agents for treatment of transplant

    rejection and auto-immune disorders.

    Mechanism of Action

    Glucocorticoids are steroid hormones that exert their physiologic actions by

    binding to the cytosolic glucocorticoid receptor.

    The glucocorticoid-glucocorticoid receptor complex translocates to the nucleus

    and binds to glucocorticoid response elements (GREs) in the promoter region of

    specific genes, either up-regulating or down-regulating gene expression.

    Glucocorticoids have important metabolic effects on essentially all cells of the

    body and, in pharmacologic doses, suppress the activation and function of innate

    and adaptive immune cells.

    Glucocorticoids lyse and induce the redistribution of lymphocytes, causing a

    rapid transient decrease in peripheral blood lymphocyte counts.

    Glucocorticoids down-regulate the expression of many inflammatory mediators,

    including key cytokines such as TNF-, interkeukin-1 (IL-1), and IL-4.

    Glucocorticoids curtail (Reduce in extent or quantity) activation of NF-B, which

    increases apoptosis of activated cells.

    Pro-inflammatory cytokines such as IL-1 and IL-6 are downregulated.

    T cells are inhibited from making IL-2 and proliferating.

    The activation of cytotoxic T lymphocytes is inhibited.

    Pharmacokinetics

    Prednisone is converted to active prednisolone in the body and has multiple

    effects on the immune system.

    Prednisone is very well absorbed from the gastrointestinal (GI) tract and has a

    long biologic half-life, so it can be dosed once daily.

    Dosing and Administration

    An intravenous corticosteroid, commonly high-dose methylprednisolone, is given

    during the perioperative period.

    The dose of methylprednisolone is tapered rapidly and discontinued within days,

    and oral prednisone is initiated.

  • 9

    Sumanth Dept of Pharmacology

    Prednisone doses are tapered progressively over time, depending on the type of

    additional immunosuppression and organ function.

    As doses are tapered, it is preferable to administer corticosteroids every other day

    and between 7 AM and 8 AM to mimic the bodys diurnal release of cortisol

    Adverse Effects

    Long-term glucocorticoid administration has important adverse effects.

    Diabetes

    Osteoporosis

    Cataracts

    Hirsutism

    Bruising

    Acne

    Hypertension

    Bone growth suppression

    Ulcerative esophagitis

    Reduced resistance to infections

    Sodium and water retention

    Increased appetite leading to weight gain

    Abrupt cessation of glucocorticoid therapy can result in acute adrenal insufficiency because the hypothalamus and pituitary gland require a number of weeks to months to re-establish adequate ACTH production.

    Therapeutic Uses

    They are combined with other immunosuppressive agents to prevent and treat transplant rejection.

    High dose pulses of intravenous Methylprednisolone sodium succinate are used to reverse acute transplant rejection and acute exacerbations of selected autoimmune disorders.

    Glucocorticoids also are efficacious for treatment of graft-versus-host disease in bone-marrow transplantation.

    Glucocorticoids are used routinely to treat autoimmune disorders such as rheumatoid and other arthritides, systemic lupus erythematosus, systemic dermatomyositis, psoriasis and other skin conditions, asthma and other allergic disorders, inflammatory bowel disease, inflammatory ophthalmic diseases, autoimmune hematologic disorders, and acute exacerbations of multiple sclerosis.

    In addition, glucocorticoids limit allergic reactions that occur with other immunosuppressive agents and are used in transplant recipients to block first-dose cytokine storm caused by treatment with muromonab-CD3 and to a lesser extent ATG (Antithymocyte Globulin).

  • 10

    Sumanth Dept of Pharmacology

    Cytotoxic Agents

    Cytotoxic Agents are used for immunosuppression as well as for antineoplastic chemotherapy. Two classes of cytotoxic agents, Antimetabolites and Alkylating agents are commonly used as Immunosuppressants.

    Antimetabolites are structural analogues of natural metabolites that inhibit essential pathways involving these metabolites.

    Alkylating agents interfere with DNA replication and gene expression by conjugating alkyl groups to DNA.

    The therapeutic goal in both antineoplastic chemotherapy and immunosuppressants is the elimination of undesirable cells.

    Antimetabolites

    They have powerful suppressive effect on immune cells. Also accompanied by many adverse effects related to their lack of selectivity.

    The older antimetabolites such as Azathioprine and Methotrexate, affect all rapidly dividing cells and can have damaging effects on the GI mucosa and Bone marrow.

    Newer antimetabolites, such as Mycophenolate mofetil and Leflunomide, cause fewer adverse effects.

    Mycophenolate mofetil mat also be more selective for immune cells, further reducing its toxicity.

    Antimetabolites typically affect both cell-mediated and humoral immunity, rendering patients more susceptible to infection.

    Azathioprine

    Azathioprine (AZA) is a prodrug for 6-mercaptopurine and has been used as an immunosuppressant in combination with corticosteroids.

    It is an imidazolyl derivative of 6-mercaptopurine. Although mercaptopurine can also be used directly as a cytotoxic agent,

    azathioprine has greater oral bioavailability and a longer duration of action and is more immunosuppressive than mercaptopurine.

    Azathioprine and mercaptopurine appear to produce immunosuppression by interfering with purine nucleic acid metabolism at steps that are required for the wave of lymphoid cell proliferation that follows antigenic stimulation.

    Mechanism of Action

    Azathioprine is an inactive compound that is converted rapidly to 6-mercaptopurine (6-MP) in the blood.

    Following exposure to nucleophiles such as glutathione, azathioprine is cleaved to 6-mercaptopurine and is subsequently metabolized by three different enzymes.

    Xanthine oxidase, found in the liver and GI tract, converts 6-MP to the inactive final end product, 6-thiouric acid.

  • 11

    Sumanth Dept of Pharmacology

    Thiopurine S-methyltransferase, found in hematopoietic tissues and red blood cells, methylates 6-MP to an inactive product, 6-methylmercaptopurine.

    Finally, hypoxanthine-guanine phosphoribosyltransferase is the first step responsible for converting 6-MP to 6-thioguanine nucleotides (6-TGNs), the active metabolites, which are incorporated into nucleic acids, ultimately disrupting both the salvage and de novo pathways of DNA, RNA, and protein synthesis. This process is toxic to the cell and renders the cell unable to proliferate.

    6-TGNs eventually are catabolized by xanthine oxidase and thiopurine S-methyltransferase to inactive products.

    Pharmacokinetics

    Oral bioavailability of azathioprine is approximately 40%. Metabolism of 6-MP is primarily by xanthine oxidase to inactive metabolites,

    which are excreted by the kidneys. The half-life of azathioprine, the parent compound, is very short, approximately

    12 minutes. The half-life of 6-MP is longer, ranging from 0.7 to 3 hours.

    However, it is the activity of the 6-TGNs that determines the pharmacodynamic half-life of the drug.

    The half-life of 6-TGNs has been estimated to be 9 days. Initial doses of azathioprine are 3 to 5 mg/kg per day intravenously or orally.

    Adverse Effects

    Dose-limiting adverse effects of azathioprine are often hematologic.

  • 12

    Sumanth Dept of Pharmacology

    Leukopenia, anaemia, and thrombocytopenia can occur within the first few weeks of therapy and can be managed by dose reduction or discontinuation of azathioprine.

    Other common adverse effects include nausea and vomiting, which can be minimized by taking azathioprine with food.

    Alopecia, hepatotoxicity and pancreatitis are less-common adverse effects of azathioprine; they generally are reversible on dose reduction or discontinuation.

    DrugDrug Interactions

    Allopurinol inhibits xanthine oxidase and can increase the bioavailability of azathioprine and 6-MP concentrations by as much as fourfold. Doses of azathioprine should be reduced by 50% to 75% when allopurinol is added.

    Therapeutic Uses

    Beneficial in maintaining renal allografts and may be of value in transplantation of other tissues.

    It is indicated as an adjunct for prevention of organ transplant rejection and in severe rheumatoid arthritis.

    Lower initial doses (1 mg/kg per day) are used in treating rheumatoid arthritis. Have been used in the management of acute glomerulonephritis and in the renal

    component of systemic lupus erythematosus. Have also proved useful in some cases of Crohns disease, and multiple sclerosis.

    Mycophenolate mofetil

    Mycophenolate mofetil is the 2-morpholinoethyl ester of Mycophenolic acid (MPA).

    Mycophenolate mofetil (MMF) is a semisynthetic derivative of Mycophenolic acid, isolated from the mold Penicillium glaucus.

    Mycophenolate mofetil is a prodrug that is rapidly hydrolysed to the active drug, Mycophenolic acid (MPA), a selective, non-competitive, and reversible inhibitor of Inosine monophosphate dehydrogenase (IMPDH), an important enzyme in the de novo pathway of guanine nucleotide synthesis.

    Mechanism of Action

    The immunosuppressive effect of MPA is exerted through non-competitive binding to Inosine monophosphate dehydrogenase, the key enzyme responsible for guanosine nucleotide synthesis via the de novo pathway.

    Inhibition of Inosine monophosphate dehydrogenase results in decreased nucleotide synthesis and diminished DNA polymerase activity, ultimately reducing lymphocyte proliferation.

    The actions of MPA are more specific for T and B cells, which use only the de novo pathway for nucleotide synthesis.

    Other cells within the body have a salvage pathway by which they can synthesize nucleotides, making them less susceptible to the actions of MPA and thereby

  • 13

    Sumanth Dept of Pharmacology

    reducing, but not eliminating, the potential for the hematologic adverse effects seen with azathioprine, which affects both the de novo and salvage pathways.

    In addition to the decreasing lymphocyte proliferation, MPA also may downregulate activation of lymphocytes.

    Selectivity

    Two main factors contribute to this selectivity.

    First, lymphocytes are dependent on the de novo pathway of purine synthesis, whereas most other tissues rely heavily on the salvage pathway. Because IMPDH is required for de novo synthesis of guanosine nucleotides but not for the salvage pathway, MPA affects only cells such as lymphocytes that rely on de novo purine synthesis.

    Second, IMPDH is expressed in two isoforms, type I and type II. MPA preferentially inhibits type II IMPDH, the isoform expressed mainly in lymphocytes.

    Together, these factors confer on MPA and MMF selectivity against T and B cells, with relatively low toxicity to other cells.

    Inhibition of IMPDH by MPA reduces intracellular guanosine levels and elevates intracellular adenosine levels, with many downstream effects on lymphocyte activation and activity.

    MPA has a cytostatic effect on lymphocytes, but can also induce apoptosis of activated T cells leading to the elimination of reactive clones of proliferative cells.

    Because guanosine is required for some glycosylation reactions, the reduction in guanosine nucleotides leads to decreased expression of adhesion molecules that are required for recruitment of several immune cell types to sites of inflammation.

    Furthermore, because guanosine is a precursor of tetrahydrobiopterin (BH4), which regulates inducible nitric oxide synthase (iNOS), the reduction in guanosine levels leads to decreased NO production by neutrophils.

    Endothelial NOS (eNOS), which controls vascular tone and is regulated by Ca2+ and calmodulin, is not affected by changes in guanosine levels, again demonstrating the considerable selectivity of MPA.

    Pharmacokinetics

    Because MPA has low oral bioavailability, it is usually administered as a sodium salt or in its prodrug form, Mycophenolate mofetil (MMF), both of which have greater oral bioavailability.

    Because MPA is unstable in an acidic environment, Mycophenolate mofetil acts as a prodrug that is readily absorbed from the GI tract, after which it is rapidly and completely converted to MPA by first-pass metabolism.

    The enteric coating of Mycophenolate sodium protects MPA from the acidic gastric pH and allows for MPA to be released directly into the small intestine for absorption.

    The absolute bioavailability of MPA when delivered from Mycophenolate mofetil and Mycophenolate sodium is 94% and 72%, respectively.

  • 14

    Sumanth Dept of Pharmacology

    Peak concentrations of MPA are reached within 1 hour following administration of either preparation.

    A total of 97% of MPA is bound to albumin in the blood. MPA is eliminated by the kidney and also undergoes glucuronidation in the liver

    to an inactive glucuronide metabolite (MPAG) that is excreted in the bile and urine.

    The half-life of MPA is 18 hours. Mycophenolate mofetil is currently available in both oral and intravenous

    formulations.

    Adverse Effects

    The most common side effects are related to the GI tract, including nausea, vomiting, diarrhea, and abdominal pain.

    Mycophenolate also has hematologic effects, such as leukopenia and anaemia, particularly with higher doses.

    Because peripheral intravenous Mycophenolate administration is associated with local edema and inflammation, central venous administration may be the preferred route.

    Therapeutic Uses

    Mycophenolate mofetil is indicated for prophylaxis of transplant rejection, and it typically is used in combination with glucocorticoids and a calcineurin inhibitor, but not with azathioprine.

  • 15

    Sumanth Dept of Pharmacology

    Its antiproliferative properties make it the first-line drug for preventing or reducing chronic allograft vasculopathy in cardiac transplant recipients.

    Mycophenolate mofetil is used as prophylaxis for and treatment of both acute and chronic graft-versus-host disease in hematopoietic stem cell transplant patients.

    Newer immunosuppressant applications for MMF include lupus nephritis, rheumatoid arthritis, inflammatory bowel disease, and some dermatologic disorders.

    Animal models show that chronic rejection is also reduced more effectively in recipients treated with MMF than in those treated with AZA or cyclosporine. The efficacy of MMF in treating chronic rejection may be related to its inhibition of both the lymphocyte and the smooth muscle cell proliferation characteristic of chronic rejection.

    Leflunomide

    Activated lymphocytes both proliferate and synthesize large quantities of cytokines and other effector molecules, and these processes require increased DNA and RNA synthesis. Therefore, agents that reduce intracellular nucleotide pools have effects on these activated cells.

    Leflunomide is an inhibitor of pyrimidine synthesis, specifically blocking the synthesis of uridylate (UMP) by inhibiting Dihydroorotate dehydrogenase (DHOD).

    DHOD is a key enzyme in the synthesis of UMP, which is essential for the synthesis of all pyrimidines.

    Experimentally, Leflunomide has been shown to be most effective in reducing B-cell populations, but a significant effect on T cells has also been observed.

    Pharmacokinetics

    Leflunomide undergoes significant enterohepatic circulation, resulting in a pro-longed pharmacologic effect.

    If Leflunomide must be removed quickly from a patients system, Cholestyramine may be administered. By binding to bile acids, Cholestyramine interrupts the enterohepatic circulation and causes a wash-out of Leflunomide.

    Adverse Effects

    The most significant adverse effects of leflunomide are diarrhea and reversible alopecia.

    Therapeutic Uses

    Leflunomide is currently approved for use in rheumatoid arthritis, but the drug has also shown significant efficacy in the treatment of other immune diseases, including systemic lupus erythematosus and myasthenia gravis.

    Leflunomide prolongs transplant graft survival and limits GVHD in animal models.

  • 16

    Sumanth Dept of Pharmacology

    Inhibition of pyrimidine synthesis by

    Leflunomide

    De novo pyrimidine synthesis

    depends on the oxidation of

    Dihydroorotate to orotate, a

    reaction that is catalysed by

    Dihydroorotate dehydrogenase.

    Leflunomide inhibits

    Dihydroorotate dehydrogenase and

    thereby inhibits pyrimidine

    synthesis.

    Because lymphocytes are

    dependent on de novo pyrimidine

    synthesis for cell proliferation and

    clonal expansion after immune cell

    activation, depletion of the

    pyrimidine pool inhibits

    lymphocyte expansion.

    Experimentally, Leflunomide

    appears to inhibit preferentially the

    proliferation of B cells; the reason

    for this preferential action is

    unknown.

  • 17

    Sumanth Dept of Pharmacology

    Cyclophosphamide

    Cyclophosphamide is a highly toxic drug that alkylates DNA.

    Cyclophosphamide has a major suppressive effect on B-cell proliferation but can enhance T-cell responses, the use of Cyclophosphamide in immune diseases is limited to disorders of humoral immunity, particularly systemic lupus erythematosus.

    Another use under consideration for Cyclophosphamide is the suppression of antibody formation against xenotransplant grafts.

    Adverse effects

    Adverse effects of Cyclophosphamide are severe and widespread, including leukopenia, cardiotoxicity, alopecia, and an increased risk of cancer because of mutagenicity.

    The risk of bladder cancer is especially notable because Cyclophosphamide produces a carcinogenic metabolite, acrolein, which is concentrated in the urine.

    When high-dose Cyclophosphamide is administered by intravenous infusion, acrolein can be detoxified by co-administration of Mesna (a sulfhydryl-containing compound that neutralizes the reactive moiety of acrolein).

    Specific Lymphocyte-Signaling Inhibitors

    Calcineurin Inhibitors (Cyclosporine and Tacrolimus)

    They are structurally unrelated and bind to distinct molecular targets, they inhibit normal T-cell signal transduction essentially by the same mechanism.

    These drugs bind to an Immunophilin (Cyclophilin for Cyclosporine or FKBP-12 for Tacrolimus), resulting in subsequent interaction with calcineurin to block its phosphatase activity.

    Calcineurin-catalyzed dephosphorylation is required for movement of a component of the Nuclear factor of activated T lymphocytes (NFAT) into the nucleus.

    NFAT, in turn, is required to induce a number of cytokine genes, including that for interleukin-2 (IL-2), a prototypic T-cell growth and differentiation factor.

    Mechanism of Action

    Inhibits the production of IL-2 by activated T-cells.

    Activated T-cells increase their production of IL-2 via a pathway that begins with dephosphorylation of a cytoplasmic transcription factor, NFAT (Nuclear factor of activated T lymphocytes).

    NFAT is dephosphorylated by the cytoplasmic Phosphatase Calcineurin. Upon dephosphorylation, NFAT translocates to the nucleus and enhances

    transcription of the IL-2 gene.

  • 18

    Sumanth Dept of Pharmacology

    Both Cyclosporine and Tacrolimus bind to Immunophilins (Cyclophilin and FK506-binding protein [FKBP], respectively), forming a complex that binds the phosphatase calcineurin.

    Calcineurin phosphatase activity is inhibited after physical interaction with the Drug-Immunophilin complex.

    This prevents NFAT dephosphorylation such that NFAT does not enter the nucleus, gene transcription is not activated, and the T lymphocyte fails to respond to specific antigenic stimulation.

    Cyclosporine also increases expression of transforming growth factor- (TGF-), a potent inhibitor of IL-2-stimulated T-cell proliferation and generation of cytotoxic T lymphocytes (CTLs).

  • 19

    Sumanth Dept of Pharmacology

    Cyclosporine

    It is a cyclic decapeptide isolated from a soil fungus, Tolypocladium inflatum.

    Pharmacokinetics

    Cyclosporine can be administered intravenously or orally. The intravenous preparation is provided as a solution in an ethanol-

    polyoxyethylated castor oil vehicle that must be further diluted in 0.9% sodium chloride solution or 5% dextrose solution before injection.

    The oral dosage forms include soft gelatin capsules and oral solutions. Cyclosporine supplied in the original soft gelatin capsule absorbed slowly with 20% to 50% bioavailability.

    After oral administration of cyclosporine, the time to peak blood concentrations is 1.5 to 2 hours.

    Administration with food delays and decreases absorption. Cyclosporine is extensively metabolized in the liver by CYP3A4 and to a lesser

    degree by the gastrointestinal tract and kidneys. At least 25 metabolites have been identified in human bile, faeces, blood, and

    urine. Only 0.1% of drug is excreted unchanged in urine. Cyclosporine and its metabolites are excreted principally through the bile into the

    faeces, with only about 6% being excreted in the urine. In the presence of hepatic dysfunction, dosage adjustments are required.

    Adverse Effects

    The principal adverse reactions to cyclosporine therapy are Nephrotoxicity Tremor Hirsutism Hypertension Hyperlipidemia

    Hyperglycaemia Gum hyperplasia Hyperuricemia Hypercholesterolemia Hepatotoxicity

    Nephrotoxicity occurs in the majority of patients treated and is the major indication for cessation or modification of therapy.

    Drug Interactions

    Substances that inhibit the enzyme CYP3A4 can decrease cyclosporine

    metabolism and increase blood concentrations. These include

    Ca2+ channel blockers (e.g., Verapamil, Nicardipine)

    Antifungal agents (e.g., Fluconazole, Ketoconazole)

    Antibiotics (e.g., Erythromycin)

    Glucocorticoids (e.g., methylprednisolone)

    HIV-protease inhibitors (e.g., Indinavir) and

    Other drugs (e.g., Allopurinol, Metoclopramide)

  • 20

    Sumanth Dept of Pharmacology

    Drugs that induce CYP3A activity can increase cyclosporine metabolism and

    decrease blood concentrations. Such drugs include

    Antibiotics (e.g., Nafcillin, Rifampin)

    Anticonvulsants (e.g., Phenobarbital, Phenytoin)

    Therapeutic Uses

    Cyclosporine may be used alone or in combination with other

    immunosuppressants, particularly glucocorticoids.

    It has been used successfully as the sole immunosuppressant for cadaveric

    transplantation of the kidney, pancreas, and liver, and it has proved extremely

    useful in cardiac transplantation as well.

    In combination with methotrexate, cyclosporine is a standard prophylactic

    regimen to prevent graft-versus-host disease after allogeneic stem cell

    transplantation.

    Cyclosporine has also proved useful in a variety of autoimmune disorders,

    including uveitis, rheumatoid arthritis, and psoriasis.

    An ophthalmic preparation of Cyclosporine is approved for the treatment of

    chronic dry eyes.

    Dosing

    The usual intravenous dose of cyclosporine is 2 to 5 mg/kg per day, given as a

    continuous infusion or, more commonly, as a single or twice-daily injection.

    Initial oral cyclosporine doses range from 8 to 18 mg/kg per day divided into two

    doses administered every 12 hours.

    Tacrolimus

    Tacrolimus is a macrocyclic triene isolated from the soil bacterium Streptomyces

    tsukubaensis.

    Pharmacokinetics

    Tacrolimus is available for oral administration as capsules and as a sterile solution

    for injection (5 mg/ml).

    Gastrointestinal absorption is incomplete and variable.

    Food decreases the rate and extent of absorption.

    Plasma protein binding of Tacrolimus is 75% to 99%, involving primarily albumin

    and 1-acid glycoprotein.

    Its half-life is about 12 hours.

    Tacrolimus is extensively metabolized in the liver by CYP3A, with a half-life of

    ~12 hours; at least some of the metabolites are active.

    The bulk of excretion of the parent drug and metabolites is in the feces.

  • 21

    Sumanth Dept of Pharmacology

    Less than 1% of administered Tacrolimus is excreted unchanged in the urine.

    Adverse Effects

    Nephrotoxicity

    Neurotoxicity (Tremor, Headache, Motor disturbances, Seizures)

    GI complaints

    Hypertension

    Hyperkalaemia

    Hyperglycaemia and Diabetes are all associated with Tacrolimus

    Drug Interactions Same as Cyclosporine

    Therapeutic Uses

    Tacrolimus is indicated for the prophylaxis of solid-organ allograft rejection.

    A topical formulation is used for the treatment of atopic dermatitis and other

    eczematous diseases

    Dosing

    Starting dose for Tacrolimus injection is 0.03 to 0.05 mg/kg per day as a

    continuous infusion.

    Recommended initial oral doses are 0.15 to 0.2 mg/kg per day for adult kidney

    transplant patients, 0.1 to 0.15 mg/kg per day for adult liver transplant patients,

    and 0.15 to 0.2 mg/kg per day for paediatric liver transplant patients in two

    divided doses 12 hours apart.

    mTOR Inhibitors

    (Sirolimus, Everolimus, Zotarolimus)

    These are the newest class of immunosuppressive agents which consists of drugs that

    target the mammalian target of Rapamycin and are collectively known as mTOR

    inhibitors.

    Sirolimus

    Sirolimus (which is also referred as Rapamycin) is a macrocyclic lactone produced

    by Streptomyces hygroscopicus.

    Sirolimus is structurally similar to Tacrolimus but have different mechanism of

    action.

    Both bind to FKBP (FK506-binding protein 12), but the Sirolimus-FKBP complex

    does not inhibit calcineurin; instead, it blocks the IL-2 receptor signalling required

    for T-cell proliferation.

  • 22

    Sumanth Dept of Pharmacology

    Mechanism of Action

    Sirolimus inhibits T-lymphocyte activation and proliferation downstream of the IL-2

    and other T-cell growth factor receptors.

    IL-2 receptor signal transduction involves a complex set of proteinprotein

    interactions that lead to increased translation of selected mRNAs encoding

    proteins required for T-cell proliferation.

    Specifically, IL-2 receptor activation initiates an intracellular signaling cascade

    that leads to phosphorylation of the molecular target of rapamycin (mTOR).

    mTOR is a serine-threonine Kinase that phosphorylates and thereby regulates the

    activity of PHAS-1 and p70 S6 kinase (P70 S6 kinase and PHAS-1 regulate

    translation).

    PHAS-1 inhibits the activity of a factor (eIF4E) required for translation, and p70 S6

    kinase phosphorylates proteins involved in protein synthesis.

    The net effect of mTOR activation is to increase protein synthesis, thereby

    promoting the transition from G1 to S phase of the cell cycle.

    Sirolimus (also known as rapamycin) crosses the plasma membrane and binds to

    intracellular FK-binding protein (FKBP).

    (Sirolimus-FKBP-12 complex does not affect calcineurin activity)

    The SirolimusFKBP complex inhibits mTOR, thereby inhibiting translation and

    causing T cells to arrest in G1.

  • 23

    Sumanth Dept of Pharmacology

    Pharmacokinetics

    Bioavailability after oral administration is low, only 15%, with peak

    concentrations being reached within 1 to 2hours.

    Sirolimus has a high volume of distribution, readily distributing into most tissues

    of the body, even though it binds extensively to erythrocytes because of the high

    FKBP12 concentration found in red blood cells.

    About 40% of sirolimus in plasma is protein bound, especially to albumin.

    Metabolism occurs primarily by CYP3A4 both in the gut and in the liver.

    The half-life is reported to be 60 hours but can be as long as 110 hours in patients

    with liver dysfunction.

    Adverse Effects

    Associated with a dose-dependent increase in serum cholesterol and triglycerides

    (Hyperlipidaemia) [mechanism of this adverse effect is related to an

    overproduction of lipoproteins or inhibition of lipoprotein lipase]

    Hyperlipidaemia

    Myelosupression

    Thrombocytopenia

    Leukopenia

    Anaemia

    GI effects

    (No Nephrotoxicity)

    Drug Interactions

    Since sirolimus is a substrate for CYP3A4 and is transported by P-glycoprotein,

    close attention to interactions with other drugs that are metabolized or

    transported by these proteins is required.

    Therapeutic Uses

    Sirolimus is indicated for prophylaxis of organ transplant rejection in combination

    with a calcineurin inhibitor and glucocorticoids.

    It is used as prophylaxis and as therapy for steroid-refractory acute and chronic

    graft-versus-host disease in hematopoietic stem cell transplant recipients.

    Topical sirolimus is also used in some dermatologic disorders and, in combination

    with cyclosporine, in the management of uveoretinitis.

    Recently, sirolimus-eluting coronary stents have been shown to reduce restenosis

    and other adverse cardiac events in patients with severe coronary artery disease.

    Dosing

    A fixed sirolimus dosing regimen is approved for concomitant use with cyclosporine

    that includes a loading dose of 6- or 15-mg followed by 2 or 5 mg daily.

  • 24

    Sumanth Dept of Pharmacology

    Cytokine Inhibition

    Cytokines (Greek cyto-cell; and kinos-movement) are small cell-signaling protein

    molecules that are secreted by numerous cells and are a category of signaling

    molecules used extensively in intercellular communication.

    The term "cytokine" has been used to refer to the immunomodulating agents, such

    as interleukins and interferons.

    Cytokines are critical signaling mediators in immune function.

    Cytokines are also pleiotropic; that is, they exert different effects depending on the

    target cell and overall cytokine milieu.

    For this reason, pharmacologic uses of cytokines or cytokine inhibitors may have

    unpredictable effects.

    The first anticytokine agent approved for clinical use was Etanercept, an anti-

    TNF- drug developed for rheumatoid arthritis.

    During the initial clinical studies, some patients with severe, drug-refractory

    rheumatoid arthritis literally got up from their wheelchairs and walked after

    receiving Etanercept.

    This dramatic efficacy ushered in a new age of biological therapies for

    autoimmune disease, and the number of new drugs that inhibit proinflammatory

    cytokines continues to grow rapidly.

    TNF- Inhibitors

    Tumor necrosis factor- (TNF- ) is a cytokine central to many aspects of the

    inflammatory response.

    Macrophages, mast cells, & activated TH cells (especially TH1 cells) secrete TNF-.

    TNF- stimulates macrophages to produce cytotoxic metabolites, thereby

    increasing phagocytic killing activity.

    TNF- also stimulates production of acute-phase proteins, has pyrogenic effects,

    and fosters local containment of the inflammatory response.

    Some of these effects are indirect and are mediated by other cytokines induced by

    TNF-.

    TNF- has been implicated in numerous autoimmune diseases.

    Rheumatoid arthritis, psoriasis, and Crohns dis-ease are three disorders in which

    inhibition of TNF- has demonstrated therapeutic efficacy.

    Proposed roles for Tumor Necrosis Factor in Rheumatoid Arthritis:

    Tumor necrosis factor (TNF) is secreted by activated macrophages in an affected

    joint, where this cytokine has multiple proinflammatory effects.

    First, TNF activates endothelial cells to up-regulate their expression of cell surface

    adhesion molecules (shown as projections on endothelial cells) and undergo other

    phenotypic changes that promote leukocyte adhesion and diapedesis.

  • 25

    Sumanth Dept of Pharmacology

    Second, TNF has a positive feedback effect on nearby monocytes and

    macrophages, promoting their secretion of cytokines such as IL-1. In turn, IL-1

    activates T cells (among other functions), and the combination of IL-1 and TNF

    stimulates synovial fibroblasts to increase their expression of matrix

    metalloproteases, prostaglandins (especially PGE 2), and cytokines (such as IL-6)

    that degrade the joint cartilage. Synovial fibroblasts also secrete IL-8, which

    promotes neutrophil diapedesis.

    Five therapies interfering with TNF- activity have been approved.

    Etanercept, Infliximab, Certolizumab pegol, Adalizumab, Golimumab

    Although all of these agents target TNF-, Etanercept is somewhat less specific

    because it binds to both TNF- and TNF-.

    Infliximab, Adalimumab, Certolizumab, and Golimumab are specific for TNF-

    and do not bind TNF- .

  • 26

    Sumanth Dept of Pharmacology

    Etanercept soluble TNF receptor dimer that links the extracellular, ligand-binding domain of human TNF receptor type II to the Fc domain of human immunoglobulin G1 (IgG1)

    Infliximab partially humanized mouse monoclonal antibody against human TNF-

    Adalimumab Fully human IgG1 monoclonal antibody against TNF- Certolizumab pegol

    pegylated anti-TNF- monoclonal antibody fragment that lacks the Fc portion of the antibody; as a result, unlike infliximab and Adalimumab, Certolizumab does not cause antibody-dependent cell- mediated cytotoxicity or fix complement in vitro.

    Golimumab fully human IgG1 monoclonal antibody against TNF- that has a longer half-life than the other anti-TNF- agents

    Etanercept

    Etanercept is a fusion protein consisting of two p75-soluble TNF receptors linked

    to an Fc fragment of human IgG1.

    MOA: The drug binds to TNF, making it biologically inactive and preventing it

    from interacting with the cell-surface TNF receptors that would lead to cell

    activation.

    The drug is given by subcutaneous injection, 50 mg once weekly or 25 mg twice

    weekly, usually through self-injections or administration by a caregiver.

    Adverse Effects

    Aside from local injection-site reactions, adverse effects are rare.

    There are case reports of pancytopenia and neurologic demyelinating syndromes

    like multiple sclerosis associated with use of Etanercept, but these are rare.

    Patients with multiple sclerosis should avoid use of this drug.

    Therapeutic Uses

    Etanercept is approved for use in

    Rheumatoid arthritis

    Juvenile idiopathic arthritis

    Plaque psoriasis

    Psoriatic arthritis

    Ankylosing spondylitis

    Infliximab

    Infliximab is a chimeric antibody combining portions of mouse and human IgG1.

    An anti-TNF antibody was created by exposing mice to human TNF.

    The binding portion of that antibody was fused to a human constant-region IgG1

    to reduce the antigenicity of the foreign protein.

  • 27

    Sumanth Dept of Pharmacology

    This antibody, when injected in humans, binds to TNF and prevents its interaction

    with TNF receptors on inflammatory cells. (Mechanism)

    Infliximab is given by intravenous infusion at a dose of 3 mg/kg at 0, 2, and 6

    weeks and then every 8 weeks.

    To prevent the formation of antibodies to this foreign protein, Methotrexate

    should be given orally in doses typically used to treat rheumatoid arthritis for as

    long as the patient continues on infliximab.

    Adverse Effects

    An acute infusion reaction with symptoms including fever, chills, pruritus, and

    rash may occur during infusion or within 1 to 2 hours after giving the drug.

    Treatment includes slowing infusion rates and administering acetaminophen,

    diphenhydramine, or corticosteroids, depending on the severity of symptoms.

    The drug may increase risk of infection.

    Autoantibodies and lupus-like syndrome also have been reported.

    Therapeutic Uses

    Infliximab is currently approved for use in

    Crohns disease

    Ulcerative colitis

    Rheumatoid arthritis

    Ankylosing spondylitis

    Plaque psoriasis

    Psoriatic arthritis

    Adalimumab

    Adalimumab is a human IgG1 antibody to TNF.

    Because it has no foreign protein components, it is less antigenic than Infliximab.

    MOA

    Like the other anti-TNF- biologicals, Adalimumab blocks the interaction of TNF-

    with TNF receptors on cell surfaces; it does not bind TNF-.

    Adalimumab lyses cells expressing TNF- in the presence of complement.

    Therapeutic Uses

    Adalimumab is approved for use in

    Rheumatoid arthritis

    Juvenile idiopathic arthritis

    Psoriatic arthritis

    Ankylosing spondylitis

    Plaque psoriasis

    Crohns disease

  • 28

    Sumanth Dept of Pharmacology

    Certolizumab pegol

    Certolizumab pegol is a recombinant humanized Fab fragment that binds to TNF-

    . It is coupled to a 40-kDa polyethylene glycol.

    It neutralizes the activity of membrane-associated and soluble TNF- without

    lysing cells.(Mechanism)

    Certolizumab is indicated for patients with

    Crohns disease

    Rheumatoid arthritis

    Golimumab

    Golimumab is a human IgG monoclonal antibody that also binds to soluble and

    membrane-associated TNF-.

    It is an intact human IgG1 and, like certolizumab pegol, it does not lyse cells

    expressing membrane-associated TNF-.

    It is indicated for patients with

    Rheumatoid arthritis

    Ankylosing spondylitis

    Psoriatic arthritis

    It has the advantage of increased half-life such that subcutaneous injections may

    be self-administered only once per month.

    General considerations for TNF- Inhibitors:

    It is important to note that high levels of TNF- are likely mediators of underlying

    pathophysiologic processes.

    However, although treatment with an anti-TNF- agent often improves disease

    symptoms, it may not reverse the underlying pathophysiology.

    Etanercept, Infliximab, Adalimumab, Certolizumab, and Golimumab are proteins

    and must be administered parenterally.

    A number of important adverse effects must be considered when administering

    TNF inhibitors.

    All patients should undergo screening for tuberculosis before initiating therapy

    because of increased risk of reactivating latent tuberculosis.

    Any patient developing an infection while taking a TNF- inhibitor should

    undergo evaluation and aggressive antibiotic treatment.

    Orally active inhibitors of TNF- and inhibitors of TNF- converting Enzyme (TACE)

    are under investigation.

  • 29

    Sumanth Dept of Pharmacology

    IL-12/IL-23p40 Inhibitors

    New biological therapies for the treatment of T-cell-mediated diseases include

    antibodies to IL-12 and IL-23.

    IL-12 and IL-23 are cytokines involved in natural killer cell activation and CD4+ T-

    cell differentiation and activation.

    IL-12, a heterodimer composed of p40 and p35 subunits, directs the differentiation

    of nave T cells into TH1 cells, which secrete IL-2, IFN-, and TNF-.

    IL-23 is also a heterodimer that has the same p40 subunit covalently linked to a

    p19 sub-unit. IL-23 directs the differentiation of nave T cells into TH17 cells, which

    secrete IL-17 and IL-22.

    Ustekinumab

    Ustekinumab is a high-affinity IgG1 human monoclonal antibody that binds to the

    p40 subunit shared by IL-12 and IL-23.

    MOA

    Ustekinumab is a human IgG1 monoclonal antibody that binds to the p40 subunit

    of IL-12 and IL-23 cytokines.

    It blocks IL-12 and IL-23 from binding to their receptors, therefore inhibiting

    receptor-mediated signaling in lymphocytes.

    Therapeutic Uses

    Ustekinumab is indicated for patients with moderate to severe plaque psoriasis

    and is in late-stage clinical trials for the treatment of multiple sclerosis and

    Psoriatic arthritis

    The advantage of Ustekinumab over anti-TNF-drugs for psoriasis is faster and

    longer term improvement in symptoms along with very infrequent dosing.

    Adverse effects include an increased risk of infections.

    IL-1 Inhibitors

    Anakinra, Rilonacept, Canakinumab

    Interleukin-1(IL-1) is an ancient cytokine, expressed in both vertebrates and

    invertebrates, that serves as a bridge between innate and adaptive immunity.

    Two forms of IL-1, IL-1 and IL-1, are encoded on different genes.

    In humans, IL-1 has primarily an immune role, while IL-1 may be involved in

    maintenance of epithelial cell function.

    Most IL-1 is generated by activated mononuclear cells.

    IL-1 stimulates IL-6 production, enhances adhesion molecule expression, and

    stimulates cell proliferation.

    IL-1 is very important in the pathogenesis of rheumatoid arthritis.

  • 30

    Sumanth Dept of Pharmacology

    It stimulates release of chemotactic factors and adhesion molecules, and these

    promote migration of inflammatory leukocytes to tissues.

    It also causes release of factors known to dilate blood vessels and direct cytotoxins

    that produce connective tissue damage.

    Modulation of IL-1 activity in vivo is accomplished in part by an endogenous IL-1

    receptor antagonist (IL-1RA)

    Anakinra

    Anakinra, a recombinant, non-glycosylated form of IL-1RA, is approved for use in

    Rheumatoid arthritis.

    Mechanism of Action

    Prevents IL-1 from binding to its receptor.

    Anakinra has modest effects on pain and swelling but significantly reduces bony

    erosions, possibly because it decreases osteoclast production and blocks IL-1

    induced metalloproteinase release from synovial cells.

    A number of rare syndromes that are mediated in part by increased levels of IL-1,

    including Muckle-Wells syndrome and Hibernian fever, have also been treated

    effectively with Anakinra.

    Collectively, these syndromes are termed Cryopyrin-associated periodic fever

    syndromes (CAPS a group of rare inherited inflammatory diseases associated

    with overproduction of IL-1 that includes Familial cold Auto inflammatory and

    Muckle-Wells Syndromes).

    Adverse Effects

    Neutropenia

    Increase susceptibility to infections

    It can be used alone or in combination with anti-TNF agents for Rheumatoid arthritis.

    Most rheumatologists feel Anakinra has a less-robust response than the TNF inhibitors

    and reserve it for use in patients who fail these agents.

    Rilonacept

    Rilonacept is a recombinant, soluble IL-1 receptor Fc fusion protein.

    It is a dimeric fusion protein consisting of the ligand-binding domains of the

    extracellular portions of the human interleukin-1 receptor component (IL-1RI) and

    IL-1 receptor accessory protein (IL-1RAcP) fused to the Fc portion of human IgG1.

    Approved for use in CAPS

    Rilonacept is now being evaluated in a phase3 study for Gout. (IL-1 is an

    inflammatory mediator of joint pain associated with elevated uric acid crystals)

  • 31

    Sumanth Dept of Pharmacology

    Canakinumab

    Canakinumab is a human IgG1 monoclonal antibody to IL-1.

    It binds to human IL-1 and prevents it from binding to IL-1 receptors.

    Approved by the FDA for CAPS.

    It is also being evaluated for use in COPD.

    Cytokine Receptor Antagonists

    An alternate approach to block the action of inflammatory cytokines is to target

    the cytokine receptor.

    This approach is attempted less frequently in drug development because there

    may be an increased risk of adverse effects if the antibody should have even

    minimal agonist activity.

    Tocilizumab

    Tocilizumab is recombinant humanized IgG1.

    MOA

    Tocilizumab binds to soluble and membrane-associated IL-6 receptors.

    It inhibits IL-6-mediated signaling on lymphocytes, suppressing inflammatory

    processes.

    The drug is administered every 4 weeks as an intravenous infusion.

    Therapeutic Uses

    It is indicated for treatment of patients with rheumatoid arthritis who are

    refractory to other anti-TNF- biologicals.

    It may be used alone or in combination with Methotrexate or other disease-

    modifying antirheumatic drugs.

    Patients taking Tocilizumab have the same increased risk of infection as those taking

    anti-TNF- monoclonal antibodies.

    Depletion of Specific Immune Cells

    Appropriately targeted antibodies deplete the immune system of reactive cells

    and thereby provide effective therapy for autoimmune diseases and transplant

    rejection.

    When the adaptive immune system reacts to an antigen, the resulting

    immunologic response includes the clonal expansion of cells specifically reactive

    against that antigen.

  • 32

    Sumanth Dept of Pharmacology

    Treatment with exogenous antibodies against cell-surface molecules that are

    expressed selectively on reactive immune cells can preferentially deplete the

    immune system of these reactive cells.

    Polyclonal Antibodies

    Antithymocyte Globulin (ATG)

    Antithymocyte globulin is a purified gamma globulin from the serum of rabbits or

    Horses immunized with human Thymocytes.

    The rabbit or horse antibodies are polyclonal and target many antigens on human

    T cells.

    Because ATG targets essentially all T cells and leads to profound lymphocyte

    depletion.

    Mechanism of Action

    Antithymocyte globulin contains cytotoxic antibodies that bind to CD2, CD3,

    CD4, CD8, CD11a, CD18, CD25, CD44, CD45, and HLA class I and II molecules on

    the surface of human T lymphocytes.

    The antibodies deplete circulating lymphocytes by direct cytotoxicity (both

    complement and cell-mediated) and block lymphocyte function by binding to cell

    surface molecules involved in the regulation of cell function.

    Therapeutic Uses

    Antithymocyte globulin also is used for acute rejection of other types of organ

    transplants and for prophylaxis of rejection.

    ATG is approved for use in prevention or treatment of renal transplant rejection,

    and the equine-derived material is also approved for treatment of aplastic anemia.

    It is provided as a sterile, freeze-dried product for intravenous administration after

    reconstitution with sterile water.

    ATG is administered intravenously once daily for upto 28 days.

    The recommended dose for acute rejection of renal grafts is 1.5 mg/kg per day.

    Adverse Effects

    Polyclonal antibodies are xenogeneic proteins that can elicit major side effects,

    including fever and chills with the potential for hypotension.

    Premedication with corticosteroids, acetaminophen, and/or an antihistamine and

    administration of the antiserum by slow infusion (over 4 to 6 hours) into a large-

    diameter vessel minimize such reactions.

    Serum sickness and glomerulonephritis (Complexes of host antibodies with horse

    ALG may precipitate and localize in the glomeruli of the kidneys) can occur.

    Anaphylaxis is a rare event.

  • 33

    Sumanth Dept of Pharmacology

    Hematologic complications include Leukopenia and Thrombocytopenia.

    There is an increased risk of infection and malignancy, especially when multiple

    immunosuppressive agents are combined.

    Monoclonal Antibodies

    Muromonab (Anti-CD3, OKT3)

    OKT3 is a mouse monoclonal antibody against human CD3, one of the cell-surface

    signaling molecules important for activation of the T-cell receptor.

    CD3 is specifically expressed on T cells (both CD4 and CD8 cells).

    Treatment with OKT3 depletes the available pool of T cells via antibody-mediated

    activation of complement and clearance of immune complexes

    Mechanism of Action

    Muromonab binds to the chain of CD3, a monomorphic component of the T-cell

    receptor complex involved in antigen recognition, cell signaling, and proliferation.

    Antibody treatment induces rapid internalization of the T-cell receptor, thereby

    preventing subsequent antigen recognition.

    Administration of the antibody is followed rapidly by depletion and extravasation

    of a majority of T cells from the bloodstream and peripheral lymphoid organs

    such as lymph nodes and spleen.

    This absence of detectable T cells from the usual lymphoid regions is secondary

    both to cell death following complement activation and activation-induced cell

    death and to margination of T cells onto vascular endothelial walls and

    redistribution of T cells to nonlymphoid organs such as the lungs.

    Muromonab-CD3 also reduces function of the remaining T cells, as defined by

    lack of IL-2 production and great reduction in the production of multiple

    cytokines, perhaps with the exception of IL-4 and IL-10.

    Pharmacokinetics

    OKT3 has a volume of distribution of 6.5 L and half-life of about 18 hours.

    Concentrations above 0.9 mcg/mL are considered therapeutic.

    If CD3 levels begin to rise, this may signify the presence of antimurine antibodies

    antagonizing the actions of OKT3.

    Administration of Mycophenolate mofetil has been suggested to reduce the

    formation of antimurine antibodies during OKT3 administration.

    Although T-cell depletion is achieved within minutes of administration,

    resolution of rejection takes 3 to 4 days.

  • 34

    Sumanth Dept of Pharmacology

    Therapeutic Uses

    Muromonab-CD3 is indicated for treatment of acute organ transplant rejection.

    Muromonab-CD3 is approved for the treatment of acute renal allograft rejection

    and steroid-resistant acute cardiac and hepatic transplant rejection.

    Adverse Effects

    OKT3 administration is associated with significant first-dose adverse reactions.

    The Cytokine-release syndrome related to OKT3, including fever, chills, rigors,

    pruritus, and alterations in blood pressure, may occur with the first several doses.

    Methylprednisolone, acetaminophen, diphenhydramine, Indomethacin, and

    pentoxifylline have been used as premedications to prevent or minimize the severity of

    this syndrome.

    Other adverse effects include

    Capillary leak syndrome and pulmonary edema, especially in fluid

    overloaded patients.

    Aseptic meningitis is another potential complication of OKT3

    therapy. If encephalitic symptoms develop, OKT3 should be

    discontinued and appropriate care initiated.

    Other adverse effects include

    encephalopathy

    nephrotoxicity

    infection, and post-transplantation lymphoproliferative disorder

    Nausea/vomiting, diarrhea, abdominal pain,

    Malaise, Myalgias, Arthralgias, and generalized weakness.

    Potentially fatal severe pulmonary edema, acute respiratory distress syndrome,

    cardiovascular collapse, cardiac arrest, and arrhythmias have been described.

    Other toxicities associated with anti-CD3 therapy include anaphylaxis and the

    usual infections and neoplasms associated with immunosuppressive therapy.

    "Rebound" rejection has been observed when muromonab-CD3 treatment is

    stopped.

    Dosing

    OKT3 should be filtered with a 0.2-to 0.22-micron filter and then administered as

    an intravenous push over 1 minute.

    The dose of OKT3 usually is 5 mg/day for 5 to 14 days.

  • 35

    Sumanth Dept of Pharmacology

    Rituximab

    Rituximab is a chimeric murine-human monoclonal IgG1 (human Fc) that binds to

    the CD20 molecule on normal and malignant B lymphocytes.

    CD20 is expressed on the surface of all mature B cells, and administration of

    Rituximab causes profound depletion of circulating B cells.

    Mechanism of Action

    The mechanism of action includes complement-mediated lysis, antibody-

    dependent cellular cytotoxicity, and induction of apoptosis in the malignant

    lymphoma cells.

    Rituximab is a monoclonal chimeric antibody consisting of mostly human protein

    with the antigen-binding region derived from a mouse antibody to CD20 protein

    found on the cell surface of mature B lymphocytes.

    The binding of Rituximab to B cells results in nearly complete depletion of

    peripheral B cells, with a gradual recovery over several months.

    Therapeutic Uses

    It is also approved for the treatment of rheumatoid arthritis in combination with

    Methotrexate in patients for whom anti-TNF- therapy has failed.

    Approved for the therapy of patients with relapsed or refractory low-grade or

    follicular B-cell non-Hodgkins lymphoma and chronic lymphocytic leukemia.

    Recent reports indicate that rituximab may also be very useful in auto-immune

    diseases such as multiple sclerosis and systemic lupus erythematosus.

    Several additional anti-CD20 antibodies are in clinical development:

    Ofatumumab

    A fully human anti-CD20 monoclonal antibody that recognizes an epitope distinct

    from that of Rituximab.

    Ofatumumab is approved for use in Chronic lymphocytic leukemia.

    Daclizumab and Basiliximab

    Daclizumab and Basiliximab are monoclonal antibodies against CD25, the high-

    affinity IL-2 receptor.

    IL-2 mediates early steps in T-cell activation.

    Because CD25 is expressed only on activated T cells, anti-CD25 antibody therapy

    selectively targets T cells that have been activated by an MHC-antigen stimulus.

    Basiliximab is a chimeric mouse-human IgG1 that binds to CD25, the IL-2 receptor

    alpha chain on activated lymphocytes.

    Daclizumab is a humanized IgG1 that also binds to the alpha subunit of the IL-2

    receptor.

  • 36

    Sumanth Dept of Pharmacology

    MOA: Both agents function as IL-2 antagonists, blocking IL-2 from binding to activated

    lymphocytes, and are therefore immunosuppressive.

    Therapeutic Uses

    Daclizumab is administered prophylactically in renal transplantation to inhibit

    acute organ rejection.

    It is also used as a component of general immunosuppressive regimens after

    organ transplantation.

    Daclizumab is typically administered in a five-dose regimen, with the first

    administration immediately after transplantation and then four additional doses

    at 2-week intervals.

    This type of dosing regimen, in which drug is administered for a limited period

    immediately after transplantation, is referred to as Induction therapy.

    Alemtuzumab

    Alemtuzumab is a humanized IgG1 with a kappa chain that binds to CD52 found

    on normal and malignant B and T lymphocytes, NK cells, monocytes,

    macrophages, and a small population of granulocytes.

    Campath-1 (CD52) is an antigen expressed on most mature lymphocytes and on

    some lymphocyte precursors.

    Its effects on depleting both T and B lymphocytes make it useful in solid-organ

    transplants.

    Mechanism of Action

    Alemtuzumab is a humanized monoclonal antibody directed at cells that express

    the CD52 surface antigen, which is found on both T and B lymphocytes, as well as

    macrophages, monocytes, eosinophils, and natural killer cells.

    When Alemtuzumab binds to the CD52 surface antigen, antibody-dependent lysis

    occurs, which removes both T and B lymphocytes from the blood, bone marrow,

    and organs, resulting incomplete lymphocyte depletion.

    Therapeutic Uses

    Alemtuzumab is approved for use in B-cell chronic lymphocytic leukemia.

    Adverse Effects

    The adverse effects of alemtuzumab include infusion-related reactions, hematologic

    effects, and infections.

    Infusion-related reactions include rigors, hypotension, fever, shortness of breath,

    bronchospasms, and chills. The potential for developing these reactions can be

    reduced by administering premedications, including corticosteroids,

  • 37

    Sumanth Dept of Pharmacology

    diphenhydramine, and acetaminophen, or by administering smaller doses and

    escalating the dose gradually.

    Hematologic effects include pancytopenia, neutropenia, thrombocytopenia, and

    lymphopenia.

    Dosing

    Commonly recommended dosing strategy for Alemtuzumab is 30 mg as a single dose.

    LFA-3

    LFA-3(also called CD58) is the counter-receptor for CD2, an antigen expressed at

    high levels on the surface of memory effector T cells.

    Interaction of CD2 on T cells with LFA-3 on antigen-presenting cells promotes

    increased T-cell proliferation and enhanced T-cell-dependent cytotoxicity.

    Because the memory effector T-cell population is elevated in patients with

    psoriasis, a pharmacologic agent (Alefacept) that disrupts the CD2LFA-3

    interaction was tested for use in psoriasis.

    Alefacept

    Alefacept is an LFA-3(leukocyte-function-associated antigen-3)/Fc fusion protein

    [Alefacept is an engineered protein consisting of the CD2-binding portion of

    leukocyte-function-associated antigen-3 (LFA-3) fused to a human IgG1 Fc

    region]that interrupts CD2LFA-3 signaling by binding to T-cell CD2, and thereby

    inhibits T-cell activation.

    Additionally, the Fc portion of Alefacept may activate NK cells to deplete the

    immune system of memory effector T cells.

    Clinically, alefacept significantly decreases the severity of chronic plaque

    psoriasis.

    Because CD2 is expressed on other adaptive immune cells, administration of

    alefacept also causes a dose-dependent reduction in CD4 and CD8 T-cell

    populations.

    Its use is therefore contraindicated in patients with HIV, and patients taking

    alefacept may have an increased risk of serious infection.

    Alefacept therapy may also be associated with an increased risk of malignancy,

    primarily skin cancer.

  • 38

    Sumanth Dept of Pharmacology

    Inhibition of Costimulation

    Abatacept, Belatacept

    Costimulationthe requirement for multiple simultaneous signals to initiate an immune responseensures that stimulation of a single immune receptor does not

    activate a damaging immune reaction. Signal 1 provides specificity, while signal 2 is permissive, ensuring that an

    inflammatory response is appropriate.

    Regulation of costimulatory molecules is a mechanism whereby the innate

    immune system regulates the extent of an immune response.

    If antigen is presented with-out a coincident costimulatory signal (i.e., without

    innate immune activation), then anergy results, whereby a cell becomes

    unreactive and will not respond to further antigenic stimuli.

    Drugs that induce anergy could be therapeutically attractive, because such agents could

    allow long-term acceptance of an organ graft or limit the extent of an autoimmune

    disease.

    Abatacept

    Abatacept consists of CTLA-4 (Cytotoxic T-Lymphocyte Antigen 4) fused to an IgG1

    constant region.

    Mechanism of Action

    Abatacept complexes with costimulatory B7 molecules on the surface of antigen-

    presenting cells.

    When the antigen-presenting cell interacts with a T cell, MHC:antigenTCR

    interaction (signal 1) occurs, but the complex of B7 with abatacept prevents

    delivery of a costimulatory signal (signal 2), and the T cell develops anergy or

    undergoes apoptosis.

    By this mechanism, Abatacept therapy appears to be effective in down-regulating

    specific T-cell populations.

    Therapeutic Uses

    Abatacept is approved for the treatment of rheumatoid arthritis that is refractory to

    methotrexate or TNF- inhibitors.

    Adverse Effects

    The major adverse effects of Abatacept are

    Exacerbations of bronchitis in patients with pre-existing

    obstructive lung disease

    Increased susceptibility to infection

  • 39

    Sumanth Dept of Pharmacology

    Abatacept should not be administered concurrently with TNF- inhibitors or anakinra

    because the combination carries an unacceptably high risk of infection.

    Belatacept

    Belatacept is a close structural congener of Abatacept that has increased affinity

    for B7-1 and B7-2.

    In a large clinical trial, Belatacept was as effective as cyclosporine at inhibiting

    acute rejection in renal transplant recipients.

    Belatacept is currently under further investigation as an immunosuppressant for

    organ transplantation.

    Blockade of Cell Adhesion

    The recruitment and accumulation of inflammatory cells at sites of inflammation

    is an essential element of most autoimmune diseases; the only exceptions to this

    rule are autoimmune diseases that are purely humoral, such as myasthenia gravis.

    Drugs that inhibit cell migration to sites of inflammation may also inhibit antigen

    presentation and cytotoxicity, thus providing multiple potential mechanisms of

    beneficial action.

    Natalizumab

    Alpha-4 integrins are critical to immune-cell adhesion and homing.

    The 4 1 integrin mediates immune-cell interactions with cells expressing

    vascular cell adhesion molecule 1 (VCAM-1), while the 4 7 integrin mediates

    immune-cell binding to cells expressing mucosal addressin cell adhesion molecule

    1 (MAdCAM-1).

    Natalizumab is a monoclonal antibody against 4 integrin that inhibits immune-cell

    interactions with cells expressing VCAM-1 or MAdCAM-1.

    Natalizumab was approved for the treatment of relapsing multiple sclerosis.

    During post marketing surveillance of the drug, however, several patients treated

    with Natalizumab developed progressive multifocal leukoencephalopathy (PML),

    a rare demyelinating disorder caused by infection with JC virus.

    This finding resulted in voluntary withdrawal of the drug.

    After further FDA investigation, it was decided to resume testing of Natalizumab

    and to add a warning to the product label regarding the possible association.

    Natalizumab was subsequently reapproved for use in the treatment of multiple

    sclerosis and Crohns disease.

  • 40

    Sumanth Dept of Pharmacology

    Inhibition of Complement Activation

    The complement system mediates a number of innate immune responses.

    Recognition of foreign proteins or carbohydrates leads to sequential activation of

    complement proteins and eventual assembly of the membrane attack complex, a

    multiprotein structure that can cause cell lysis.

    Patients with paroxysmal nocturnal hemoglobinuria (PNH) have acquired defects

    in complement regulatory proteins, leading to inappropriate activation of

    complement and complement-mediated lysis of erythrocytes.

    Eculizumab

    Eculizumab is a humanized monoclonal antibody against C5, a complement

    protein that mediates late steps in complement activation and triggers assembly of

    the membrane attack complex.

    Eculizumab is approved for the treatment of PNH; it significantly decreases

    hemoglobinuria and the need for erythrocyte transfusions in patients with this

    disorder.

    Genetic evidence indicates that complement activation may play an etiologic role

    in age-dependent macular degeneration, suggesting that inhibitors of the

    complement cascade could be useful local therapies for this disease.

    Immunostimulants

    In contrast to immunosuppressive agents that inhibit the immune response in transplant

    rejection and autoimmunity, a few immunostimulatory drugs have been developed with

    applicability to infection, immunodeficiency, and cancer.

    Levamisole

    Thalidomide

    Bacillus Calmette-Guerin (BCG)

    Recombinant Cytokines (Interferons)

    Interleukin-2

    Levamisole

    Levamisole was synthesized originally as an anthelmintic but appears to "restore"

    depressed immune function of B lymphocytes, T lymphocytes, monocytes, and

    macrophages.

    In combination with the antimetabolite 5-fluorouracil, this drug is now approved

    for use in the treatment of colon cancer.

  • 41

    Sumanth Dept of Pharmacology

    Thalidomide

    Thalidomide is best known for the severe, life-threatening birth defects it caused

    when administered to pregnant women.

    For this reason, it is available only under a restricted distribution program and

    can be prescribed only by specially licensed physicians who understand the risk

    of teratogenicity if thalidomide is used during pregnancy.

    Thalidomide should never be taken by women who are pregnant or who could

    become pregnant while taking the drug.

    It is indicated for the treatment of patients with erythema nodosum leprosum and

    also is used in conditions such as multiple myeloma.

    Its mechanism of action is unclear. Thalidomide has been reported to decrease

    circulating TNF- in patients with erythema nodosum leprosum.

    Alternatively, it has been suggested that the drug affects angiogenesis.

    The anti-TNF- effect has led to its evaluation as a treatment for severe, refractory

    rheumatoid arthritis.

    Bacillus Calmette-Guerin (BCG)

    Live bacillus Calmette-Guerin is an attenuated, live culture of the bacillus of

    Calmette and Guerin strain of Mycobacterium bovis that induces a granulomatous

    reaction at the site of administration.

    By unclear mechanisms, this preparation is active against tumors and is indicated

    for treatment and prophylaxis of carcinoma in situ of the urinary bladder and for

    prophylaxis of primary and recurrent stage Ta and/or T1 papillary tumors after

    transurethral resection.

    Adverse effects include hypersensitivity, shock, chills, fever, malaise, and immune

    complex disease.

    Recombinant Cytokines-Interferons

    Although interferons (alpha, beta, and gamma) initially were identified by their

    antiviral activity, these agents also have important immunomodulatory activities.

    The interferons bind to specific cell-surface receptors that initiate a series of

    intracellular events:

    induction of certain enzymes

    inhibition of cell proliferation

    enhancement of immune activities, including increased

    phagocytosis by macrophages

    augmentation of specific cytotoxicity by T lymphocytes.

    Recombinant interferon alfa-2b (IFN-alpha 2, INTRON A)

    Obtained from Escherichia coli by recombinant expression.

  • 42

    Sumanth Dept of Pharmacology

    It is a member of a family of naturally occurring small proteins with molecular

    weights of 15,000 to 27,600 daltons, produced and secreted by cells in response to

    viral infections and other inducers.

    Therapeutic Uses

    Interferon alfa-2b is indicated in the treatment of a variety of tumors, including

    hairy cell leukemia, malignant melanoma, follicular lymphoma, and AIDS-related

    Kaposi's sarcoma.

    It also is indicated for infectious diseases, chronic hepatitis B, and condylomata

    acuminata.

    In addition, it is supplied in combination with ribavirin (REBETRON) for

    treatment of chronic hepatitis C in patients with compensated liver function not

    treated previously with interferon alfa-2b or who have relapsed after interferon

    alfa-2b therapy.

    Adverse Effects

    Flu-like symptoms, including fever, chills, and headache, are the most common

    adverse effects after interferon alfa-2b administration.

    Adverse experiences involving the cardiovascular system (hypotension,

    arrhythmias, and rarely cardiomyopathy and myocardial infarction) and CNS

    (depression, confusion) are less-frequent side effects.

    Interferon gamma-1b (ACTIMMUNE)

    Is a recombinant polypeptide that activates phagocytes and induces their

    generation of oxygen metabolites that are toxic to a number of microorganisms.

    It is indicated to reduce the frequency and severity of serious infections associated

    with chronic granulomatous disease.

    Adverse reactions include fever, headache, rash, fatigue, GI distress, anorexia,

    weight loss, myalgia, and depression.

    Interferon beta-1a (AVONEX, REBIF) a 166-amino acid recombinant glycoprotein

    Interferon beta-1b (BETASERON), a 165-amino acid recombinant protein

    Have antiviral and immunomodulatory properties.

    They are FDA approved for the treatment of relapsing and relapsing-remitting

    multiple sclerosis to reduce the frequency of clinical exacerbations.

    The mechanism of their action in multiple sclerosis is unclear.

    Flu-like symptoms (fever, chills, myalgia) and injection-site reactions have been

    common adverse effects.

  • 43

    Sumanth Dept of Pharmacology

    Interleukin-2

    Human recombinant interleukin-2 (Aldesleukin, PROLEUKIN; des-alanyl-1,

    serine-125 human IL-2) is produced by recombinant DNA technology in E. coli.

    This recombinant form differs from native IL-2 in that it is not glycosylated, has

    no amino terminal alanine, and has a serine substituted for the cysteine at amino

    acid 125.

    The potency of the preparation is represented in International Units in a

    lymphocyte proliferation assay such that 1.1 mg of recombinant IL-2 protein

    equals 18 million International Units.

    Aldesleukin has the following in vitro biologic activities of native IL-2:

    Enhancement of lymphocyte proliferation and growth of IL-2-

    dependent cell lines;

    Enhancement of lymphocyte-mediated cytotoxicity and killer cell

    activity; and induction of interferon-g activity.

    In vivo administration of Aldesleukin in animals produces multiple immunologic

    effects in a dose-dependent manner.

    Cellular immunity is profoundly activated with lymphocytosis, eosinophilia,

    thrombocytopenia, and release of multiple cytokines (e.g., TNF, IL-1, and

    interferon-g).

    Therapeutic Uses

    Aldesleukin is indicated for the treatment of adults with metastatic renal cell

    carcinoma and melanoma.

    Adverse Effects

    Administration of Aldesleukin has been associated with serious cardiovascular

    toxicity resulting from capillary leak syndrome, which involves loss of vascular

    tone and leak of plasma proteins and fluid into the extravascular space.

    Hypotension, reduced organ perfusion, and death may occur.

    An increased risk of disseminated infection due to impaired neutrophil function

    also has been associated with Aldesleukin treatment.

    Tretinoin

    Tretinoin, or all-trans retinoic acid (ATRA), is a ligand of the retinoic acid receptor

    (RAR). ATRA is used in the treatment of acute promyelocytic leukemia.

    This disease is characterized by a translocation t(15;17) in which part of the RAR

    gene is fused to the PML gene, creating a fusion protein that induces a block to

    differentiation and thereby allows development of the leukemia.

    Treatment with ATRA stimulates differentiation of these cells into more normal

    granulocytes.

  • 44

    Sumanth Dept of Pharmacology

    In some patients, the induction of differentiation can lead to a life-threatening

    over-production of white blood cells.

    ATRA can also induce a rapidly progressive syndrome of fever, acute respiratory

    distress with pulmonary infiltrates, edema and weight gain, and multisystem

    organ failure.

    Therapy with high doses of glucocorticoids is often an effective treatment for this

    ATRA syndrome.

    References

    Laurence Brunton, Bruce Chabner, Bjorn Knollman, Goodman & Gilmans

    The Pharmacological Basis Of Therapeutics, 12th Edition, Mc Graw Hill

    Medical, 2011 Pg No: 1005-1023

    David E.Golan, Armen H.Tashjian, Ehrin J. Armstrong, April W. Armstrong,

    Principles of Pharmacology, The Pathophysiologic Basis Of Drug

    Therapy,3rd Edition, Lippincott Williams & Wilkins, 2012 Pg no: 790-806

    H.P.Rang, M.M. Dale, J.M. Ritter, R.J. Flower, Rang and Dales

    Pharmacology, 6th Edition, Churchill Livingstone Elsevier, 2007

    Bertram G. Katzung, Susan B. Masters, Anthony J. Trevor, Basic & Clinical

    Pharmacology, 12th Edition, Tata McGraw Hill Education Private Limited,

    New Delhi, 2012, Pg No: 977-999