Txs for autoimmune disease Flashcards

1
Q

What are our calcineurin inhibitors?

A
  • Cyclosporin

- Tacrolimus

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Discuss the structure of cyclosporin? (Where do we get it, what is the active component)

A

Cyclosporine is a hydrophobic cyclic peptide produced by nonribosomal peptide synthesis in the fungus Tolypocladium inflatum Gams. The ring contains a novel 9-carbon amino acid at position 1 that is essential for biological activity, as are the residues involved in the formation of intramolecular hydrogen bonds

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Principal effects of cyclosporin and mechanism of action?

A

The principal effect is to inhibit the activation and proliferation of CD4+, and CD8+ T-cells.

These effects are the result of inhibition of transcription of cytokine encoding genes (e.g., IL-2). Within the cell, cyclosporine binds with high affinity to the intracellular protein cyclophilin. The cyclosporinecyclophilin complex is a potent inhibitor of calcineurin, a phosphoprotein phosphatase.

Inhibition of this phosphatase blocks the nuclear translocation of a transcription factor (NF-ATc) required for the expression of the IL-2 gene.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Discuss the weird pharmacology of cyclosporin

A

Alright so this guy is water insoluble. We need to administer it in a vehicle containing EtOH and oil when giving it IV, and if we must give it orally, put it in a gelatinous capsule.

Metabolism is mediated by CYP3A4, so we see increased clearence with phenytoin, phenobarbital, or really anything that induces P-450.

Inhibitors of CYP3A4 such as erythromycin, ketoconazole, amphotericin B and St. John’s wart help increase activity (as you might expect)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Toxicity of cyclosporins?

A

nephrotoxicity, neurotoxicity (tremor, seizure), hypertension, hirsutism, hyperlipidemia, gingival hyperplasia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Discuss the general structure of tacrolimus and what makes it better than cyclosporin

A

A macrocylic lactone-lactam antibiotic structurally unrelated to cyclosporine. It is 50-100 times more potent than cyclosporine, and has less nephrotoxicity. In addition to its use in the prevention of transplant rejection and GVHD, topical application of tacrolimus is used to treat atopic dermatitis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Mechanism of Tacrolimus?

A

Effects on immune system are virtually identical to those of cyclosporine.

However, tacrolimus primarily binds to another cytoplasmic protein (FKBP-12) to form a complex that also inhibits calcineurin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Discuss the pharmacology differences of Tacrolimus when compared to cyclosporine

A

Also metabolized in the liver;
shorter half-life than cyclosporine - thus after three day iv loading it is given orally twice daily. Used primarily for prophylaxis after kidney and liver transplants; also for rescue therapy in patients experiencing graft rejection

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Toxicities of Tacrolimus

A

Toxicities: nephrotoxicity, neurotoxicity (tremor, headache, seizures, insomnia), hypertension, inhibition of pancreatic β-cell function (i.e diabetes), and increased risk of lymphoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What the fuck is Pimecrolimus?

A

Structurally similar to tacrolimus; also binds to FKBP-12 which results in inhibition of calcineurin; used for topical treatment of atopic dermatitis - 2nd-line therapy ONLY because of increased risk of lymphoma and malignant melanoma.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What is Sirolimus (Rapamycin) and how does it differ from Tacrolimus and cyclosporine?

A

Structurally related to tacrolimus, but with an entirely different mechanism of action. It is as effective as cyclosporine in preventing acute rejection after renal transplantation.

Because it has little if any renal toxicity, it can be included with cyclosporine to prevent transplant rejection. Sirolimus is also used to prevent neointimal proliferation and restensosis after stent placement in coronary arteries

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Discuss the mechanism of action for Sirolimus (Rapamycin)

A

Like tacrolimus, rapamycin binds to FKBP-12. However, instead of inhibiting calcineurin activity, the sirolimus-FKBP complex inhibits the activity of the PI 3- kinase-related kinase protein mTOR (Mammalian Target Of Rapamycin), which modulates a variety of intracellular signaling pathways that regulate the transcription and translation of genes involved in cellular proliferation.

Inhibition of mTOR is manifested as a lack of response to growth stimulatory signals, and inhibition of T and B cell proliferation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Discuss the metabolism of Sirolimus (Rapamycin)

A

Metabolized in the liver primarily by the same enzyme (CYP3A4) responsible for the metabolism of cyclosporine and tacrolimus. Used as part of combination therapy after renal transplant, and experimentally after islet cell transplantation.

Everolimus, a sirolimus analogue, offers more predictable oral absorption.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Side effects of sirolimus (rapamycin)

A

anemia, thrombocytopenia, and hyperlipidemia; there is a higher incidence of post-surgical lymphocoele in transplant patients receiving sirolimus; contraindicated for use after liver or lung transplantation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Describe mycophenolate mofetil and what do we with it

A

a prodrug metabolized to the “antimetabolite” mycophenolic acid (MPA). Used in solid organ transplant recipients as a single agent or in combination with low dose cyclosporine or tacrolimus to reduce calcineurin-inhibitor toxicity.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Mechanism of action for mycophenolate mofetil

A

mycophenolate mofetil is hydrolyzed by liver esterases to MPA, which is a potent inhibitor of inosine monophosphate dehydrogenase, an enzyme required for de novo purine biosynthesis. Selectivity is based on the fact that in T and B cells, the major source of purines is de novo synthesis rather than HPRT (hypoxanthine guanine phosphoribosyl transferase) mediated salvage.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

When do we use mycophenolate mofetil and what do we worry about in regards to is absorption

A

recommended for use after renal and heart transplantation; should not be administered with antacids containing magnesium or aluminum hydroxide because of decreased absorption

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Side effects for mycophenolte mofetil

A

Diarrhea, leukopenia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Talk about azathioprine and what we use it for.

What toxicities do we worry about?

A

another anti-proliferation prodrug – active metabolite 6-mercaptopurine is a purine analog that disrupts de novo purine biosynthesis and inhibits DNA replication. Used in combination therapy with prednisone -/+ CsA/tacrolimus) to prevent orgran rejection, or as an adjunct DMARD for treatment of severe RA.

Toxicity = GI distress and leukopenia.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

What the hell is a Disease-Modifying Anti-Rheumatic Drug

A

DMARDs represent a mixed group of compounds with demonstrated anti-rheumatoid activity despite unrelated, and often poorly understood, mechanisms of action.

Although traditionally used as secondary agents when traditional NSAIDs fail, certain DMARDs are now utilized as “first-line” therapy (e.g. methotrexate, leflunamide, sulfasalazine) for rheumatoid arthritis upon diagnosis.

Given that clinical efficacy is delayed (i.e. months), early concomitant treatment with NSAIDs and glucocorticoids is common.

21
Q

What is the mechanism of action and general function of methotrexate (cellularly, not what we necessarily use it for, that question comes next)

A

a folic acid antagonist that allosterically inhibits dihydrofolate reductase (DHFR), the enzyme required for tetrahydrofolate production, a key component of nucleoside biosynthesis. Also appears to disrupt other enzymes required for nucleoside biosynthesis, such as AICARD transformylase.

Demonstrates cytotoxic and anti-proliferative activity on lymphocytes, which require de novo nucleoside synthesis for DNA/RNA replication. May block T cell activation due to inhibition of purine metabolism and adenosine accumulation

22
Q

What do we use methotrexate for and what do we worry about with it?

A

Commonly used as first-line therapy for moderate to severe RA or psoriasis, as well as cancer chemotherapy.

Side effects include ulcerative stomatitis and leukopenia/anemia – low-dose folic acid supplementation may ameliorate these symptoms.

Methotrexate is highly teratogenic and should not be used during pregnancy.

23
Q

Discuss the metabolism of sulfasalazine and its mechanism of action

A

a prodrug composed of a sulfonamide (sulfapyridine) and salicylate, processed by gut bacteria into 5-aminosalicylic acid. Mechanism of action is unclear, but may work by scavenging reactive oxygen species produced by neutrophils.

24
Q

What do we use sulfasalazine for? What do we worry about?

A

Relieves joint pain and swelling and induces remission in active RA. Also used for treatment of Crohn’s disease. Side effects include GI distress, leukopenia. Like other sulfonamides, sulfasalazine may trigger anaphylactic reactions in some patients.

25
Q

What do we use penicillamine for and how does it work?

A

dimethylcysteine produced by hydrolysis of penicillin. D-isomer shows antirheumatoid activity in ~75% RA patients, perhaps connected to decreased IL-1 production and collagen maturation in inflamed joints. Also used as a metal chelator for Wilson’s disease or heavy metal poisoning – cannot be combined with gold compounds

26
Q

Side effects of penicillamine?

A

Side effects include rashes, stomatitis, GI distress, and potential proteinuria, leukopenia and thrombocytopenia.

Contraindicated for patients who are pregnant or have history of renal disease

27
Q

Effects, use, and side effects for gold compounds

A

sodium aurothiomalate and auranofin can be used to reduce joint pain and swelling and slow progression of joint damage. Therapeutic effects are slow in onset (3-4 months).

Mechanism of action is unknown.

Side effects include rashes, stomatitis, proteinuria, thrombocytopenia, and potential neuropathy or hepatitis in some patients

28
Q

Effect, use, side effects for chloroquine/hydroxychloroquine

A

anti-malarial drugs often used in combination with methotrexate and sulfasalazine for treatment of mild to moderate RA.

Mechanism of action is unknown, with therapeutic efficacy only noted in ~50% patients with slow onset (2-4 months).

Ocular toxicity is a rare side effect.

29
Q

Discuss the function and side effects for leflunomide

A

a prodrug whose active metabolite inhibits dihydropteroate dehydrogenase, an essential mitochondrial enzyme in de novo pyrimidine biosynthesis.

Approved for use in treatment of active rheumatoid arthritis. Prevents expansion of activated lymphocytes, which cannot utilize the salvage pyrimidine pathway. Extremely long half-life; side effects include diarrhea, nausea, myleosuppression with blood cytopenias, and infrequent but severe hepatotoxicity.

30
Q

Mechanism of action for the exciting drug tofacitinib

A

1st approved inhibitor of Janus kinases – inhibits JAK3/JAK1 > JAK2, without affecting TYK2. Inhibits JAK/STAT signaling associated with multiple cytokines, including IFN-g, IL-6, and those that use the common gamma chain (e.g. IL-2, IL-4, IL-7, etc.).

Blocks T cell differentiation and production of pro-inflammatory mediators in joint tissue.

31
Q

What do we use tofacitinib for and what are the side effects we worry about

A

Approved for treatment of RA patients who have failed initial MTX therapy; can be used as monotherapy or in combination with MTX and other DMARDs.

Side effects include increased infections, lymphoma, neutropenia/anemia, and elevated LDL. Metabolized by CYP3A4; must be used cautiously with other drugs that affect CYP3A4 expression/activity

32
Q

Discuss monoclonal antibodies and recombinant proteins and what we tend to generally use them for

A

Monoclonal antibodies (mAbs) (and certain recombinant proteins derived from cellular receptors) are designed to specifically interact with a single target molecule, often neutralizing its activity. They are used as immunosuppressive agents in transplantation, inflammatory or autoimmune disorders

33
Q

Discuss the function and effects of the monoclonal antibody Muromonab

A

mouse monoclonal antibody directed against the ε chain of the T cell surface protein CD3; blocks engagement of the T cell receptor; once indicated for reversal of acute rejection of heart, liver, and kidney transplants.

Can non-specifically activate T cells upon first infusion to cause cytokine release syndrome, inducing an adverse, systemic inflammatory response. Hypersensitivity reactions may also occur.

34
Q

When do we use daclizumab and basiliximab and how do they work?

A

recombinant chimeric (human/mouse) monoclonal antibodies directed against the α chain (CD25) of the high-affinity IL-2 receptor; inhibits IL-2-mediated Tcell activation; reduces incidence of acute rejection when used in combination with cyclosporine/prednisone/azathioprine in kidney and cardiac transplantation.

Use of basiliximab can result in acute hypersensitivity reactions.

35
Q

Discuss etanercept

A

recombinant chimera of soluble p75-TNF receptor type II and Fc portion of human IgG; presumably acts by neutralizing free TNF. Used in treatment of rheumatoid arthritis, ankylosing spondylitis, and plaque psoriasis. Use of etanercept has been associated with increased incidence of demyelinating diseases

36
Q

Discuss anakinra

A

a recombinant nonglycosylated analog of human IL-1 receptor antagonist (IL-1RA); approved for treatment of rheumatoid arthritis and in patients with mutations in IL-1 RA gene; principal side effect is increased risk of serious bacterial infection.

37
Q

Discuss tocilizumab

A

humanized monoclonal antibody directed against IL-6 receptor that blocks the proinflammatory effects of IL-6; used to treat rheumatoid arthritis in patients unresponsive to TNF-α inhibitors

38
Q

Discuss rituximab

A

a recombinant chimeric (mouse/human) anti-CD20 monoclonal antibody that promotes complement mediated lysis of CD20-positive B cells; used to treat rheumatoid arthritis as well as non-Hodgkin’s lymphoma and chronic lymphocytic leukemia

39
Q

Discuss natalizumab

A

humanized monoclonal antibody against α4subunit of α4β1-integrin that inhibits lymphocyte migration through endothelial cell sites of inflammation; used for treatment of Crohn’s disease and multiple sclerosis. Use is associated with significant risk of JC virus induced progressive multifocal leukoencephalopathy (PML).

40
Q

Discuss abatacept. What do we use it for and what do we worry about?

A

a recombinant chimera of the extracellular domain of CTLA-4 and the Fc portion of human IgG1 used for treatment of rheumatoid arthritis; binds to CD80 and CD86 to block binding to CD28 and prevent T cell activation; works upstream from infliximab and etanercept.

41
Q

Discuss omalizumab

A

humanized monoclonal antibody that binds to immunoglobulin E (IgE); high levels of IgE are associated with allergic reactions; approved for treating moderate-to-severe allergic asthma that is unresponsive to corticosteroids.

42
Q

What the hell is fingolimod and what do we use it for?

A

fingolimod: when phosphorylated by sphingosine kinase 2, binds to sphingosine-1 phosphate receptors and blocks migration of lymphocytes out of lymph nodes; used to treat multiple sclerosis. Adverse effects associated with fatal infections.

43
Q

Discuss using thalidomide vs. Lenalidomide

A

Thalidomide is a sedative drug with immunomodulatory and anti-inflammatory properties, including inhibition of angiogenesis; used for treatment of multiple myeloma (usually in combination with dexamethasone), erythema nodosum leprosum (type 2 leprosy reactions).

Mechanism of action is unknown; toxicities include peripheral neuropathy (sometimes irreversible) and teratogenicity; should NEVER be given to pregnant women or women of childbearing potential.

Lenalidomide, an immunomodulatory derivative (IMiD) of thalidomide, is less teratogenic and approved for treatment of myeloma and myelodysplastic syndrome involving chromosome 5q31 deletion.

44
Q

What are anti-lymphocyte and anti-thymocyte globulins?

A

isolated from hyperimmune horse or rabbit serum after immunization with human thymic lymphocytes; contains antibodies to numerous T cell surface antigens; depletes peripheral T cells by blocking cell surface receptors and by direct cytotoxicity; used in combination with other immunosuppressive agents

45
Q

How does IVIG work?

A

purified, polyvalent human IgG used to replace antibodies in a variety of immunodeficiencies to confer passive immunity; also used to treat autoimmune disorders such as idiopathic thrombocytopenic purpura (ITP).

Precise mechanism of action in suppressing inflammation and autoimmunity remains controversial.

46
Q

What do we treat with IFN-B?

A

Multiple sclerosis

47
Q

What do we use epoetin alfa for?

A

recombinant erythropoeitin analogue that stimulates formation of red blood cells in anemic patients

48
Q

What do we use darbepoetin alfa for?

A

Longer half life than epoetin alfa!

Use in cancer chemotherapy induced anemia is controversial because these agents may stimulate tumor cell proliferation.

In kidney failure patients, target hemoglobin levels should be below 12 g/dL. Higher levels have increased risk of stroke and cardiovascular side effects.

Methoxy polyethylene glycolepoetin beta has a very long biological half-life.