Current and Future Oral Systemic Therapies for Psoriasis




For patients with moderate to severe psoriasis, there is a large range of variably effective and safe oral, systemic medications. With appropriate monitoring, these therapies may be used as either monotherapy or in combination with other therapies. Newer drugs in the research pipeline hold significant promise.


Key points








  • Methotrexate is a relatively safe, long-term treatment of appropriately screened and monitored patients who can be expected to achieve a 75% reduction in the Psoriasis Area and Severity Index in approximately 40% of cases.



  • For patients without any contraindications, cyclosporine in most cases is a rapidly effective, short-term medication frequently used as a bridge to longer term maintenance approaches to the control of psoriasis.



  • Carefully selected and monitored patients, especially those with the pustular, palmoplantar or erythrodermic forms of psoriasis, may derive significant benefit from acitretin therapy.



  • Apremilast (approved in the US for both psoriasis and psoriatic arthritis) and tofacitinib (approved in the US for rheumatoid arthritis) are novel oral medications that have shown effectiveness in clinical trials; these medications are likely to be approved for use in patients with moderate to severe psoriasis within the near future.



  • Mycophenolate mofetil, 6-thioguanine, leflunomide, and other oral agents remain reasonable alternatives, but various aspects of their efficacy, monitoring requirements, and adverse event profiles relegate these drugs to patients who have failed both oral and biologic medications.




Psoriasis is a chronic, immunodysregulatory disease with significant prevalence and a range of severity in its dermatologic and rheumatologic signs and symptoms. Topical agents remain the first-line treatments for mild disease. For patients with greater percent body surface area (BSA) of involvement, or with disease that has significant quality of life impact, systemic therapy or phototherapy is indicated. Strong evidence indicates that psoriasis is an independent risk factor for cardiovascular risk and that the inflammatory comorbidities of psoriasis merit systemic therapy even in the absence of joint disease, though prospective data to assess whether there is a morbidity or mortality benefit to doing so are in their infancy.


Systemic medication for psoriasis can be broadly divided into small molecules, which are usually given orally, and biologics, which are large molecules that must be delivered by injection or infusion. Systemic medications are often used as monotherapy but, in many, instances are combined or used with phototherapy to increase or maintain efficacy; decrease undesired effects; or, out of necessity, for refractory disease. In some cases oral systemics are given in combination with biologics for synergistic effects or to forestall the development of biologic immunogenicity.


The small molecules used in therapy for psoriasis include mature drugs, such as methotrexate, cyclosporine, and acitretin, as well as newer compounds discovered through targeted development and understanding of molecular pathways, such as tofacitinib and apremilast. This article focuses on the, small molecule systemic therapies with clinical data supporting their efficacy.




Patient assessment


A thorough medical history, prior treatment history and focused physical examination should be the starting point from which systemic therapy for psoriasis is considered, with a review of systems that covers the cutaneous, immune, hematologic, cardiac, gastrointestinal, musculoskeletal, neurologic, reproductive, and family and social history. Concerns of the patient regarding mode of administration and risk aversion need to be considered ( Table 1 ). This evaluation should include laboratory investigations, and it is prudent to include baseline studies necessary for the whole spectrum of possible systemic treatments. A suggested list of these tests is included in Table 2 . A complete picture of the patient’s disease severity, impact on their quality of life, overall health and comorbidities, and their willingness to attempt available treatment should coalesce, and a patient-centered, mutually agreeable therapeutic approach enacted. The nonbiologic, nonphototherapy options for modern systemic treatment of psoriasis are as follows.



Table 1

Selected patient considerations in systemic psoriasis therapy


































Parameter Consideration
Extent or severity of psoriasis Typically >10% BSA, or involved areas significantly impede quality of life
Presence of psoriatic arthritis Consider systemic, disease-modifying therapy regardless of level of skin involvement
Woman of childbearing potential Not a good candidate for most oral psoriasis medications
Man attempting to conceive a child Not a good candidate for methotrexate
Chronic or binge alcohol user Avoid concomitant therapies with significant risk of hepatotoxicity
History of hepatitis Avoid therapies with significant risk of hepatotoxicity
History of hematologic malignancy Use immunosuppressants with caution
Immunodeficiency Avoid immunosuppressants
Smoker Counsel on quitting


Table 2

Suggested baseline laboratory workup before systemic psoriasis therapy




























Frequent Less Frequent
Comprehensive metabolic panel Magnesium, phosphate
Complete blood count β-human chorionic gonadotropin
HIV serology
Lipid panel
Hepatitis B serology
Hepatitis C serology
Interferon-gamma releasing assay for the detection of latent tuberculosis or purified protein derivative tuberculin




Patient assessment


A thorough medical history, prior treatment history and focused physical examination should be the starting point from which systemic therapy for psoriasis is considered, with a review of systems that covers the cutaneous, immune, hematologic, cardiac, gastrointestinal, musculoskeletal, neurologic, reproductive, and family and social history. Concerns of the patient regarding mode of administration and risk aversion need to be considered ( Table 1 ). This evaluation should include laboratory investigations, and it is prudent to include baseline studies necessary for the whole spectrum of possible systemic treatments. A suggested list of these tests is included in Table 2 . A complete picture of the patient’s disease severity, impact on their quality of life, overall health and comorbidities, and their willingness to attempt available treatment should coalesce, and a patient-centered, mutually agreeable therapeutic approach enacted. The nonbiologic, nonphototherapy options for modern systemic treatment of psoriasis are as follows.



Table 1

Selected patient considerations in systemic psoriasis therapy


































Parameter Consideration
Extent or severity of psoriasis Typically >10% BSA, or involved areas significantly impede quality of life
Presence of psoriatic arthritis Consider systemic, disease-modifying therapy regardless of level of skin involvement
Woman of childbearing potential Not a good candidate for most oral psoriasis medications
Man attempting to conceive a child Not a good candidate for methotrexate
Chronic or binge alcohol user Avoid concomitant therapies with significant risk of hepatotoxicity
History of hepatitis Avoid therapies with significant risk of hepatotoxicity
History of hematologic malignancy Use immunosuppressants with caution
Immunodeficiency Avoid immunosuppressants
Smoker Counsel on quitting


Table 2

Suggested baseline laboratory workup before systemic psoriasis therapy




























Frequent Less Frequent
Comprehensive metabolic panel Magnesium, phosphate
Complete blood count β-human chorionic gonadotropin
HIV serology
Lipid panel
Hepatitis B serology
Hepatitis C serology
Interferon-gamma releasing assay for the detection of latent tuberculosis or purified protein derivative tuberculin




Methotrexate


Methotrexate is the oldest systemic therapy for moderate to severe psoriasis having been approved over 40 years ago. Its anti-inflammatory effects are primarily mediated through its metabolism to polyglutamate derivatives that are potent inhibitors of 5-aminoimidazole-4-carboxamide ribonucleotide transformylase. In so doing, methotrexate increases the endogenous levels of adenosine, a potent anti-inflammatory compound. Methotrexate also has been shown to decrease primary and secondary antibody responses. Generally, it is available in 2.5 mg tablets and is typically prescribed at 7.5 to 25 mg, taken once weekly; higher doses produce better response. Older dosing schemes were split between 3 administrations, every 12 hours, but this is unnecessary from an efficacy or side-effect standpoint. Additionally, subcutaneous methotrexate dosed once weekly has displayed better bioavailability, efficacy, and tolerability relative to the oral formulation in studies of patients with rheumatoid arthritis. Daily or weekly folic or folinic acid supplements are recommended to decrease side effects (particularly bone marrow suppression) and improve gastrointestinal tolerability.


Because methotrexate use for psoriasis predates the modern drug approval process, there have been, until the last decade, a lack of robust clinical data regarding its efficacy. This, however, is no longer the case. A multicenter, prospective, randomized, double-blind, placebo-controlled study involved 110 subjects receiving methotrexate doses from 7.5 to 25 mg/wk (plus folate 5 mg 2 days after the methotrexate dose). At 16 weeks, using the Psoriasis Area and Severity Index (PASI), 35.5% of subjects achieved a 75% reduction in PASI (PASI 75). A similar response rate, PASI 75 of 39.9% at 24 weeks, was seen in a comparison study of methotrexate and briakinumab. These results parallel the largest single study involving methotrexate, in which 41.9% of the 215 subjects taking methotrexate (in an open-label, nonblinded manner) achieved PASI 75 at 16 weeks. Although these PASI 75 response rates are somewhat lower than reported in other clinical trials, they represent the best quality data, and a reasonable expectation of oral methotrexate efficacy is that approximately 40% will achieve PASI 75. The advent of pharmacogenomics and identification of gene polymorphisms conferring a better response to methotrexate may facilitate patient screening and increase the percentage achieving optimal control, once such testing becomes more readily available.


The most common clinical side effects of methotrexate at the low doses used for psoriasis are nausea, diarrhea, fatigue, and headache. Supplementation with folic and/or folinic acid may either mitigate or eliminate these adverse effects, which tend to become less severe with time. Monitoring for rarer and more severe side effects is an integral part of every follow-up appointment. Liver function tests (LFTs) should be checked every 4 to 12 weeks because the first signs of hepatotoxicity may manifest as an elevation in transaminases. For long-term monitoring of hepatotoxicity, the general movement has been away from routine liver biopsy and toward noninvasive measurement of classic and newly validated serum biomarkers, such as type III procollagen amino terminal propeptide, which is currently standard in most European countries. In some jurisdictions, liver biopsy may be recommended before methotrexate initiation in patients with hepatic disease risk factors, and by American Academy of Dermatology (AAD) guidelines once a cumulative dose of 3.5 to 4 g is reached. Particular vigilance to hepatic monitoring is necessary because psoriasis has been identified as an independent risk factor for nonalcoholic fatty liver disease and the comorbidities often found in psoriatic patients can place them at higher risk of methotrexate-induced hepatic injury.


A complete blood count is necessary to warn of myelosuppression, whereas adequate kidney function must be verified because most methotrexate is renally excreted. Dose adjustments can be made for decreased glomerular filtration rate, but this should be done with care because these patients are at increased risk of cytopenia. Screening for any changes in pulmonary status, including breathing difficulty or persistent cough, is a requirement because of the rare side-effect of idiosyncratic pneumonitis or pulmonary fibrosis. Other adverse effects include alopecia (higher or, inadvertently, daily doses), mouth ulcers (particularly in the setting of neutropenia), and (rarely) radiation recall and/or acral erythema. In patients with rheumatoid arthritis, an increased frequency of melanoma, non-Hodgkin lymphoma and lung cancer has been reported ; however, these findings have not been well-substantiated.


Methotrexate has numerous potential interactions with other drugs. Care should be taken with concurrent salicylates, regular use and overuse of nonsteroidal anti-inflammatory drugs, and certain types of antibiotic use (particularly sulfonamides), and it is recommended to check a current interaction database before initiating new therapy.


Methotrexate is pregnancy category X; pregnancy and lactation are absolute contraindications. Individual accounts of accidental in utero exposure without any observed sequelae exist, but all patients (male and female) should discontinue methotrexate 3 months before any planned attempts at pregnancy, and those pregnancies conceived while on methotrexate should be considered high-risk until proven otherwise.


Methotrexate often is combined with other systemic therapies or phototherapy to achieve either additive or synergistic effects. Combination allows lower doses; minimized toxicity; augmented efficacy; and, via the reduction in immunogenicity, the preservation of the activity of biologic therapies. Combination may involve cyclosporine, acitretin (with careful liver monitoring, in appropriate patients), TNF-α inhibitors, and ustekinumab. The combination of methotrexate with another therapy frequently leads to superior and longer lasting efficacy than with either therapy alone.




Cyclosporine


Cyclosporine is a calcineurin inhibitor that is approved to treat moderate to severe psoriasis. Its mechanism of action is to form a complex with cyclophilin, resulting in reduced activity of the nuclear transcription factor, nuclear factor of activated T cells, thus lowering the production of IL-2 and other proinflammatory cytokines, and, consequently, inhibiting T cell activation. In addition cyclosporine inhibits interferon-γ production. At typical doses (2.5–5.5 mg/kg/d) cyclosporine is highly effective for rapidly and consistently treating severe disease. Cyclosporine therapy may be used with many different approaches: (1) intermittently as a short-course (12–16 week) therapy with minimal renal and hypertensive toxicity, (2) continuously to maintain remission in a minority of patients with refractory disease, (3) as a crisis intervention (4–8 weeks) to reduce flare or treat severe disease, and (4) as a part of sequential and rotational therapy to help minimize toxicity and optimize efficacy. The treatment regimen should be tailored to the needs and medical history of the individual patient.


Unlike methotrexate, cyclosporine has not been used as a comparator in any modern, large, multicenter, randomized, placebo-controlled clinical trials. There are, however, several moderate-size clinical trials that demonstrate the drug’s high efficacy ( Tables 3 and 4 ). Studies have established a clear dose-response curve in which 5 mg/kg/d is the inflection point that balances safety and side effects with impressive clearance; cyclosporine at this dose reliably results in average PASI 75 rates of 70%. Further, there seems to be improved benefit in starting at this dose, then titrating down as the disease is controlled.



Table 3

Select clinical studies evaluating oral methotrexate


































































































































Study, Year Agent Number of Subjects Evaluated Study Type Outcome Measure Results P Comments
Heydendael et al, 2003 MTX vs CsA 85 (43 MTX) RCT, assessor blinded PASI 75 At 16 wk MTX 15–22.5 mg/wk: 60%
CsA 3–5 mg/kg: 71%
P = .29 comparing 2 groups receiving MTX and CsA No placebo arm; doses of both MTX and CsA could be increased to achieve response; no folate supplementation in MTX arm. Treatment had to be discontinued in 12 subjects in the methotrexate group because of elevated liver enzyme levels
Flyström et al, 2008 MTX vs CsA 68 (37 MTX) MRCT PASI reduction; calculated PASI 75 At 12 wk MTX PASI 75 21.6%; CsA 3–5 mg/kg 64% P = .0028 5 mg daily folate in MTX arm; 7.5–15 mg/wk MTX
Ho et al, 2010 MTX vs Traditional Chinese Medicine vs placebo 50 (19 MTX) RPCT PASI 75 At 24 wk MTX PASI 75 63%, placebo PASI 75 18% P <.01 5 mg daily folate in MTX arm; 10–30 mg/wk MTX
Akhyani et al, 2010 MTX vs MMF 38 (18 MTX) Open label RCT PASI 75 At 12 wk MTX PASI 75 73.3%, MMF PASI 75 58.8% P >.05 1 mg folate daily (except MTX day); MTX titrated up to 20 mg/wk
Saurat et al, 2008 MTX vs adalimumab vs placebo 271 (110 MTX) MRDPCT PASI 75 At 16 wk, MTX PASI 75 35.5%; adalimumab PASI 75 79.6% P <.001 ∼5 mg folate/wk; MTX dose escalation 7.5–25 mg/wk
Barker et al, 2011 MTX vs infliximab 868 (215 MTX) Open-label RCT PASI 75 At 16 wk MTX PASI 75 41.9%; infliximab PASI 75 77.8% P <.001 Folate recommended but not mandated; dose escalation to 20 mg MTX permitted at week 6
Radmanesh et al, 2011 MTX daily vs weekly 202 (101 each daily or weekly) RCT PASI 75 At 16 wk: daily PASI 75 60%, weekly PASI 75 81% P <.001 No placebo arm; no folate supplements; no topical or other systemic therapy. increased nausea/vomiting/fatigue in weekly group
Dogra et al, 2012 MTX 10 or 25 mg/wk 51 (25 at 10, 26 at 25 mg/wk) RDCT PASI 75 At 12 wk, 10 mg PASI 75 72%; 25 mg PASI 75 92% P >.05 Folate 5 mg twice each week; excluded subjects with body mass index >30
Baranauskaite et al, 2012 MTX 15 mg/wk vs MTX 15 mg/wk + infliximab 94 (47 MTX) Open-label RCT PASI 75 (in those with baseline PASI >2.5) At 16 wk MTX alone PASI 75 54.3%; infliximab + MTX PASI 75 97.1% P <.0001 More adverse events with dual therapy; no comment on folate supplementation
Ranjan et al, 2007 MTX vs hydroxyurea 30 (15 MTX) Open-label RCT PASI 75 At 12 wk, MTX PASI 75 66.7%, hydroxyurea PASI 75 13.3% Weekly dosing of hydroxyurea; less adverse events than reported with daily dosing
Mahbub et al, 2013 MTX vs hydroxyurea 24 (13 MTX) Case or control PASI reduction At 8 wk, MTX had 85% PASI reduction; hydroxyurea had 78% P >.05
Fallah Arani et al, 2011 MTX vs fumarates 51 (26 MTX) RCT PASI 75 At 16 wk MTX PASI 75 42%; fumarates PASI 75 38% P >.65 MTX dose-escalated up to 15 mg/wk; folate used
Reich et al, 2011 MTX vs briakinumab 317 (163 MTX) MRDPCT PASI 75 At 24 wk, MTX PASI 75 39.9%, briakinumab PASI 75 81.8% P <.001 95/163 MTX discontinued study due to lack of efficacy; folate 5 mg/wk

Abbreviations: CsA, cyclosporine; MMF, mycophenolate mofetil; MRCT, multicenter, randomized clinical trial; MRDPCT, multicenter, randomized, double-blind, placebo-controlled clinical trial; MTX, methotrexate; NA, not available; RCT, randomized clinical trial; RDCT, randomized, double-blind clinical trial; RPCT, randomized, placebo-controlled clinical trial.

Data from Refs.


Table 4

Select additional clinical studies evaluating cyclosporine


























































Study, Year Agent Number of Subjects Evaluated Study Type Outcome Measure Results P Comments
Ellis et al, 1991 CsA (2.5 –7.5 mg/kg/d) vs placebo 85 RDPCT PGA and Global Severity Score At 8 wk, 36%, 65%, and 85% were clear or almost clear on doses of 2.5 mg/kg/d, 5 mg/kg/d, or 7.5 mg/kg/d, respectively P <.0001 Higher doses associated with more renal toxicity, hypertension, intolerance
Reitamo et al, 2001 CsA 34 RDPCT PASI improvement At 16 wk, CsA 1.25 mg/kg 33%; CsA 5 mg/kg 71% P <.001
Yoon & Youn, 2007 CsA 61 Open-label PASI 75 At 12 wk, 75% of step-down subjects and 51.5% of standard subjects achieved PASI 75 P <.05 Standard regimen 2.5 mg/kg/d titrated up vs step-down regimen of 5 mg/kg/d titrated down
Pedraz et al, 2006 CsA vs MMF 8 Sequential PASI improvement At 16 wk CsA PASI 75 85%, MMF PASI 75 50% NA CsA 4 mg/kg/d, MMF 30 mg/kg/d
Beissart et al, 2009 CsA vs MMF 37 (21 CsA) Multicenter open-label RCT PASI 75 At 12 wk CsA PASI 75 58%, MMF PASI 75 18% P <.01 2.5 mg/kg CsA
2 g/d MMF; dose escalation at 6 wk if <25% improvement in PASI

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Feb 12, 2018 | Posted by in Dermatology | Comments Off on Current and Future Oral Systemic Therapies for Psoriasis

Full access? Get Clinical Tree

Get Clinical Tree app for offline access