Biologics in Dermatology: A Brief Review

Iffat Hassan, Samia Aleem, Gousia Sheikh and Parvaiz Anwar

Cite this article as: BJMP 2013;6(4):a629
Download PDF


Abstract

With the advent of biologic therapy, the treatment of various systemic and cutaneous diseases, especially autoimmune diseases, has been revolutionized. Most of the treatment modalities available prior to biologics aimed at producing clinical improvement of the disease without targeting the actual causative factors. Biologics are protein molecules produced by recombinant DNA technology, which target the specific sites in the immune-pathogenesis pathway of the diseases. Because of the specific action on immune system, biologics are presumed to have lesser side effect profile compared to the traditional immune-suppressants. However, the use of biologics is still limited because of unknown long-term safety profile and various aspects of the biologics need to be thoroughly evaluated by conducting large scale studies worldwide. In this review we give a brief description of various biologic agents that are known till date.

Keywords: Biologics; proteins; autoimmune diseases.

INTRODUCTION

The US Food and Drug Administration (USFDA) considers the following as biologics: any therapeutic serum, toxin, antitoxin, vaccine, virus, blood, blood component or derivative, allergenic product, analogous product, or derivatives applicable to the prevention, treatment, or cure of injuries or disease of man1. However, generally, biologics refer to protein molecules therapeutically used in various diseases so as to target specific points in the inflammatory cascade of these disorders 2. Hope for an improved tolerability, convenience in usage and lasting remissions, combined with increased knowledge of immune-pathogenesis of various cutaneous diseases has lead to the introduction of biologics as alternative immune-modulating agents in the field of dermatology.

CLASSIFICATION

Biologics are generally divided into three major groups 3:

a) Monoclonal antibodies

b) Fusion antibody proteins

c) Recombinant human cytokines and growth factors

The main groups and the principal agents in each group are summarised in Box 1 and described below.

A) MONOCLONAL ANTIBODIES

Monoclonal antibodies target specific cell-surface receptors. In the early days of biologic therapy, purely murine monoclonal antibodies were used. However, due to the development of antimurine antibodies, which blocked their action, these could be given only for very short periods. The monoclonal antibodies used now have different amounts of murine sequences in the variable region. They may be categorised into three classes:

(a) chimeric antibodies comprising of 30% murine genes fused with human antibodies

(b) humanised antibodies, which have 10% murine sequences, and

(c) human antibodies, which are solely derived from human immunoglobulin genes 4.

Principal monoclonal antibodies with therapeutic relevance in Dermatology

The principal monoclonal antibodies known till date are enumerated in Box 1 and described briefly below.

Box 1: Classification of biologics
Monoclonal antibodies
    · Anti-TNFα: Infliximab, Adalimumab, Certolizumab, Golimumab
    · Anti-LFA1: Efalizumab
    · Anti-CD20: Rituximab
    · Anti-IL-12 and anti-IL-23 monoclonal antibody: Ustekinumab
    · Anti-CD2 antibody: Siplizumab
    · Anti-CD4 antibody: Orthoclone (OKTcdr4a)
    · Anti-CD25 antibodies: Basiliximab, Daclizumab
    · Anti-CD80r: Galiximab (IDEC 114)
    · Anti-IgE: Omalizumab
Fusion antibody proteins
    · Etanercept
    · Alefacept
    · Abatacept
    · Onercept
    · Denileukin Diftitox
Recombinant human cytokines and growth factors
  a) Interferons
    · Interferon α (IFNα)
    · Interferon γ (IFNγ)
    · Interleukin 1 Receptor antagonist (IL1Ra)
    · Interleukin 2 (IL-2)
    · Interleukin 4 (rhIL-4)
    · Interleukin 10 (rhIL-10)
    · Interleukin 11 (rhIL-11)
  b) Granulocyte macrophage colony stimulating factor (GM-CSF)
  c) Platelet derived growth factor (PDGF)

1. Infliximab

Infliximab (trade name Remicade) is a human-mouse monoclonal antibody that binds to and inhibits the activity of TNF-α, and also causes lysis of TNF-α producing cells5.

Important uses of Infliximab

Psoriasis: Infliximab is approved for the treatment of psoriatic arthritis and plaque psoriasis by FDA6, 7. Infliximab may also be of value in recalcitrant or unstable disease and in generalised pustular psoriasis. It is given as an IV (intravenous) infusion in doses of 5 or 10 mg/kg, over a period of 2 hours at weeks 0, 2, 6 and may be followed by repeat single infusions at 8-12 week intervals 8. In various controlled trials, improvement at 10 weeks has been noted in 87% of patients 7, 9.

Atopic dermatitis: Infliximab has also been evaluated in a study of atopic dermatitis 10. At 2 weeks, there was significant improvement in all patients. At 10, 14, and 30 weeks, variable response was seen.

Hidradenitis suppurativa: Long-term efficacy has also been evaluated in hidradenitis suppurativa. In one study, some patients had no evidence of recurrence after 2 years, while others relapsed within a mean of 8.5 months11.

2. Adalimumab

Adalimumab (Humira®) is a human IgG1 monoclonal antibody directed against TNF-α. Adalimumab is given in a dose of 40 mg subcutaneously (SC) every other week as self-injection 5, 12.

Important uses of Adalimumab

Psoriasis: Adalimumab rapidly reverses the decrease in epidermal Langerhans cell density in psoriatic plaques 13. In a trial, patients with psoriatic arthritis received adalimumab every other week for 24 week14. It was well-tolerated and helped improve joint and skin manifestations significantly.

Hidradenitis suppurativa: An increasing number of reports in refractory hidradenitis supprativa have shown successful control with adalimumab 15, 16, 17.

3. Basiliximab

Successful treatment for severe psoriasis and generalised pustular psoriasis has been reported with basiliximab, an interleukin-2 receptor (IL-2R; CD25) chimeric monoclonal antibody 18, 19.

4. Daclizumab

Daclizumab is a humanised monoclonal antibody thatbinds to the CD25 subunit of the IL-2 receptor onT-cells, thus blocking T-cell proliferation. It has been tried in recalcitrant psoriasis and HIV-associated psoriatic erythroderma with a mean reduction in PASI of 30% 20, 21 22.

5. Siplizumab

Siplizumab (Medi-507) is a humanisedmonoclonal antibody directed against CD2. It is designed to block stimulationby inhibiting the CD2–LFA-3 interaction. In earlyphase studies in psoriasis, significant response to therapy has been noted23.

6. Efalizumab

Efalizumab is a recombinant humanised monoclonal IgG1 antibody that binds to CD11a, a subunit of leukocyte function-associated antigen 1 (LFA-1) 24. It destabilises and decreases the trafficking of T-cells into dermal and epidermal tissues.

Important uses of Efalizumab

Psoriasis: Efalizumab was approved by the US FDA in October 2003 for the treatment of psoriasis5. It is currently the only biologic agent approved for continuous administration to adult patients24. The licensed dose of efalizumab is 1 mg/kg weekly as a subcutaneous self- administered injection for 12 weeks, following a first conditioning dose of 0.7 mg/kg 24.

Lichen planus: There is one case report of 3 months duration of treatment with efalizumab for lichen planus with resolution of skin lesions and pruritis 25.

7. Rituximab

Rituximab is a monoclonal humanised antibody directed again in the B cell-specific antigen CD20.

Important uses of Rituximab

Lymphoma: It has been used in patients with CD20-positive non-Hodgkin's lymphoma in a dosage of 375 mg/m2 for four infusions26.

SLE: In systemic lupus, dose escalation studies revealed no differences with respect to clinical outcome in patients who received either a single infusion of 100 mg/m2, a single infusion of 375 mg/m2, or four weekly infusions of 375 mg/m2 27.

Blistering diseases: For patients with blistering diseases, most patients receive the lymphoma dosage schedule. However, serious side effects were considerably higher.

There are reports of refractory pemphigus patients who received infusions of rituximab and had rapid resolution of lesions and a long lasting clinical remission 28, 29, 30.

8. Galiximab

Galiximab a humanised monoclonal antibody directedagainst CD80 and blocks its interaction with CD28 on the T cell, for T-cell stimulation 31. Clinicaldata for this drug are just beginning to emerge with 40% of patients achieving at least 50% reduction in PASI after receiving4 biweekly doses in a trial32, 33.

9. Ustekinumab

It is a fully human monoclonal antibody targeting IL-12 and IL-23, presently undergoing clinical trials for psoriasis and psoriatic arthropathy2, 34. In placebo-controlled studies, (PHOENIX 1) and (PHOENIX 2) have shown that ustekinumab could control plaque psoriasis with only four injections a year resulting in greater ease of use and more sustained relief 35, 36.

10. Certolizumab pegol

Certolizumab is the recombinant antibody Fab' fragment of a humanised TNF inhibitor monoclonal antibody. A study in chronic plaque psoriasis showed that certolizumab pegol, given subcutaneously every two weeks, over a period of 12 weeks shows significant improvement 37.

11. Golimumab

Golimumab, a fully human monoclonal antibody is at present undergoing Phase III clinical trials in psoriatic arthropathy 38.

12. Orthoclone or OkT4a

It is a humanised antihuman CD4 IgG4 monoclonal antibody preventing the recognition of the MHC-bound antigen by an appropriate T-cell receptor, hence T cells do not get activated 39. Several studies have found orthoclone to be effective in moderate to severe psoriasis 40, 41.

13. ABX-IL8

ABX-IL8 is a fully human monoclonalantibody designed to bind free IL-8, a key chemokine in psoriasis and deactivate it in theskin42, 43. In early trials, the drug has demonstrated good clinical responsein psoriasis 44, 45.

14. Omalizumab

Omalizumab is a recombinant, humanised, monoclonal antibody against immunoglobulin IgE. This agent acts as a neutralising antibody by binding IgE at the same site on IgE as its high-affinity receptor, FcεR I, thus inhibiting the biological effects before the generation of allergic symptoms 46. There are reports of the efficacy of omalizumab in chronic urticaria 47, cold urticaria 48 and atopic dermatitis 49.

15. Mepolizumab

Mepolizumab is a humanised monoclonal IgG antibody to the IL-5 molecule, which is essential for eosinophil growth and differentiation. Two weekly infusions showed significant clinical improvement in atopic dermatitisand clinical trials are underway for hyper-eosinophillic disorders 50, 51.

16. SMART Anti–IFN-γ

SMART anti–IFN- γ,a humanised monoclonal antibody, binds and inactivates IFN- γ, an important Th1 cytokinein psoriasis. Early phase studies are being performed at this time 52.

B) FUSION ANTIBODY PROTEINS

Fusion proteins, also known as chimeric proteins, are proteins which are created by the fusion of the receptor domain of a human protein with the constant region of human IgG. The resultant fusion protein binds specifically to a ligand or co-receptor 53. The most commonly used fusion proteins in dermatology are briefly described below and enumerated in Box 1.

1. Alefacept

Alefacept is a bivalent recombinant fusion protein composed of the first extracellular domain lymphocyte function antigen 3 (LFA-3), fused to the hinge domain of human IgG1. The LFA-3 portion of alefacept binds to CD2 receptors on T-cells, thereby blocking their natural interaction with LFA-3 on antigen presenting cells (APCs). The IgG1 portion of alefacept binds to FcγR receptor on natural killer cells to induce T-cell apoptosis 54.

Important uses of Alefacept

Psoriasis: The US FDA approved alefacept in January 2003 for treatment in adult patients with moderate to severe chronic plaque psoriasis. It is given by intramuscular or intravenous route with a dose of 10-15 mg IM weekly or 7.5 mg IV weekly and a 12 week course is recommended 5, 54. Two 12-week courses showed a75% or greater reduction in the PASI 55.

Alopecia areata: Case reports have shown that alefacept may be effective in the treatmentof AA56, 57.

Pyoderma gangrenosum: Alefacept has been used for pyoderma gangrenosum and improvement was shown in 25% of these patients 58.

Other Indications

Some of the off-label conditions where alefacept has been used with success are graft-versus-host disease (GVHD), lichen planus, alopecia areata, atopic dermatitis, mycosis fungoides,alopecia universalis, erosive lichen planus,Hailey-Hailey diseaseand hand dermatitis 58, 59, 60, 61, 62, 63.

2. Denileukin diftitox

Denileukin diftitox is a novel recombinant fusion protein consisting of fragments of diphtheria toxin linked to human interleukin-2 and works by targeting the high-affinity interleukin-2 receptors. It was tried in patients with recalcitrant psoriasis and the rate of improvement for treated patients was found to be significant 64.

3. Abatacept (Ctla4ig)

It is a fusion protein composed of the extracellular domain of CTLA4 and the Fc region of IgG4. It interferes with T-cell activation by competitively binding the B7.1 and B7.2 molecules on the surface of APC 65. In a study, patients with stable psoriasis vulgaris showed good improvement with IV infusion of abatacept 33. A second generation CTLA4Ig, Belatacept, is currently under Phase II clinical trial for allograft diseases 66.

4. Etanercept

Etanercept is a recombinant fully human dimeric fusion protein comprising of the human TNF-α p75 receptor and the Fc portion of human IgG1 molecule. It functions as a TNF inhibitor, thereby preventing interaction with its cell surface receptors on target cells and blocking its pro-inflammatory effects.

Important uses of Etanercept

Psoriasis: Etanercept (Enbrel®) is FDA approved for use as subcutaneous monotherapy in psoriasis. Several clinical trials have shown that it is effective67, 68, 69, 70. The adult dose is 50 mg/week, given subcutaneously for three months 5. The drug is also indicated for treatment of psoriatic arthritis 71, 72.

Hidradenitis supprivata:There is a study of etanercept in patients with severe hidradenitis with more than 50% score improvement12.

5. Onercept

Onercept is a recombinant human soluble p55 tumour necrosis factor binding protein under development for the potential treatment of psoriasis and psoriatic arthritis73.
C) RECOMBINANT HUMAN CYTOKINES AND GROWTH FACTORS

Cytokines are non-immunoglobulin proteins and glycoproteins produced by a wide variety of cells in the human body and released in response to any immune stimulus 74, 75. Recombinant cytokines or cytokine antagonists have been used as immunomodulators 76. The principal recombinant cytokines used in dermatology, enumerated in Box 1, are described below.

1. Interferons (IFNs)

IFNs, a family of related proteins, are produced by virus-infected leucocytes. They exhibit anti-proliferative, immune-modulatory and anti-neoplastic functions 77.

· Interferon α

Recombinant IFNα is given as a subcutaneous or intramuscular injection to treat verruca vulgaris 78 , condyloma acuminatum 79, cutaneous T cell lymphoma 80, Kaposi's sarcoma (AIDS related) 81, melanoma 82, basal cell carcinoma 83, squamous cell carcinoma 84, actinic keratosis 85, Behçet's disease 86, hemangiomas 87 and keloids 88.

The injections are usually given thrice weekly and the dose (depending on the condition being treated) varies from low-dose therapy for condyloma acuminatum to high-dose therapy for melanoma 89, 90. Of late, pegylated IFNα is being used for convenience, because it has a longer half-life and hence can be given once weekly 80.

· Interferon-γ

It is FDA approved for the treatment of chronic granulomatous disease 91 and has also been used in atopic dermatitis 92 and cutaneous T cell lymphoma 90.

2. Interleukin 1 receptor antagonist (IL1Ra, Anakinra)

Anakinra is the non-glycosylated form of human IL-1Ra and acts by blocking the functions of the naturally occurring IL-193. Good results have also been reported in Schnitzler's syndrome94, familial cold auto-inflammatory syndrome 95 and psoriatic arthropathy96. It is given by subcutaneous injection 100 mg once a day.

3. Interleukin 2

Recombinant IL-2 is an antitumour cytokine that has been used in cutaneous T cell lymphoma (CTCL) and metastatic melanoma 97. When given intravenously in high doses of 600,000-720,000 IU/kg in melanoma, IL-2 has produced a 15-20% overall response, with complete cure in 4-6% 98.

4. Interleukin 4 (rhIL-4)

In a dose-escalation study (0.5 to 5mg/kg given by subcutaneous injection thrice a week), IL-4 has been shown to cause improvement in psoriasis by inducing Th2 differentiation in human CD4 + T cells 99.

5. Interleukin 11 (rhIL-11, Oprelvekin)

It has also shown reasonably good results in the treatment of psoriasis at doses of 2.5 or 5mg/ kg, by subcutaneous injection 100.

6. Granulocyte macrophage colony stimulating factor (GM-CSF)

GM-CSF acts by stimulating stem cells to produce granulocytes, monocytes and macrophages 101. Recombinant human GM-CSF has been used to promote wound healing in ulcerated skin for example leg ulcers 102, 103, and for the treatment of melanoma 104 and Sezary syndrome 105.

7. Platelet derived growth factor (PDGF)

PDGF is a dimeric glycoprotein which regulates and promotes granulation tissue formation, re-epithelialisation and wound angiogenesis 106. Recombinant PDGF-BB topical gel (100ìg/g), applied once daily, has been approved by FDA for the treatment of diabetic foot ulcers 107, 108.

8. Recombinant Human IL-10

Recombinant Human IL-10 (Tenovil)can be given in subcutaneousinjections. Early phase clinical trials have shown thatrecombinant human IL-10 three times a week improved psoriasis 109, 110.

SIDE EFFECTS OF BIOLOGICS 5, 111, 112, 113

Some of the adverse effects of biologics are described below:

  • Allergic reaction and antibody formation: Mostly seen with TNF-α blockers.
  • Mild transient injection site reactions: Comprising of erythema, oedema and bruising, noted with etanercept in 10-20% of cases in the first month of therapy. Antibodies to etanercept may develop in 6% of patients.
  • Infusion reaction: Occurs during or within 1-2 hours of treatment and may affect up to 20% of all the patients treated with infliximab, rarely anaphylactic shock may occur.
  • Acute flu-like symptoms: Headache, chills, fever, nausea and myalgia may occur within 48 hours after administration of the first two doses of efalizumab and Interferon α.
  • Infections: Reactivation of tuberculosis may occur on treatment with anti-TNF-α agents and sepsis secondary to Listeria monocytogenes and Histoplasma capsulatum have been reported 113.
  • Malignancy: Patients previously treated with PUVA represent an at-risk group.
  • Demyelinating disease: Worsening of multiple sclerosis and demyelination reported with infliximab.
  • Thrombocytopenia: Occurs with efalizumab and warrants discontinuation of therapy.
  • Autoimmune haemolytic anemia: Occurs 4-6 months after the start of treatment with efalizumab.
  • Congestive cardiac failure: Worsening of congestive cardiac failure with TNF-α blockers is reported to occur.
  • Antinuclear antibodies and lupus-like syndrome: May develop during therapy with anti-TNF-α agents, but not associated with symptoms and signs of lupus in the majority.
  • Hepatitis: Reported following infliximab therapy, occurring from 2 weeks to more than a year after initiation of treatment. Treatment should be stopped in the event of jaundice and/ or marked elevations (>5 times the upper limit of normal) in liver enzymes.

PATIENT SCREENING FOR BIOLOGIC THERAPY 113, 114

All patients to be put on biologics should undergo a thorough evaluation including detailed clinical history, physical examination and relevant investigations with particular reference to known toxicity profile of the agent being considered. The investigations generally advised are113: full blood count, liver and renal function tests, screening for hepatitis and HIV infection, anti-nuclear antibodies, anti-ds DNA, urine analysis, chest X-ray and Tuberculin skin testing.

For efalizumab, haemogram (including platelet count) is recommended monthly for the first 3 months and then every 3 months. For TNF blockers, it is done at 3 months initially and repeated every 6 months.

Liver and renal function tests, serum electrolytes and urine analysis are done at 3 months initially and then every 6 months.

EXCLUSION CRITERIA/ CONTRAINDICATIONS

There are various contraindications for use of biologics, warranting their exclusion and precautions are to be exercised because of their immune-modulator properties. The main exclusion criteria are: active tuberculosis, severe congestive heart failure, patients having >200 treatments of PUVA (because of a risk of developing malignancies with anti- TNF agents), history of demyelinating disease or optic neuritis, hepatitis B and C positivity, HIV positivity, premalignant states, active infections and high risk states such as chronic leg ulcers, persistent or recurrent chest infections and indwelling urinary catheter infections, pregnancy and breast-feeding.

ASSESSMENT OF THE RESPONSE TO BIOLOGICS

Many scoring systems for assessing the severity of various dermatological diseases exist. These scoring systems and other indices can be used for assessment of response to the use of biologics. For example, for evaluation of improvement in psoriasis, PASI (psoriasis area and severity index) and DLQI (dermatology life quality index) are recommended at 3 months initially and then every 6 months 113. Reduction in baseline PASI score of >75% is the standard used by FDA to assess the efficacy of a new psoriasis agent 115. Similarly in atopic eczemas, improvement is monitored based on the Eczema Area and Severity Index, Pruritus Severity Assessment and DLQI. Reduction of the Eczema Area and Severity Index score by 50% is considered excellent, 30-49% moderate and <29% non-significant.

SUMMARY

To summarise, biologics represent the future of therapeutics, not only in dermatology but also in other fields of medicine. Among the various dermatological disorders where they are used, biologics have been most evaluated in psoriasis 116. However, the possibility of serious infections and the oncogenic potential combined with the high cost of the drugs limit their routine use at the present stage 117. Regular re-evaluation of efficacy and safety is essential if these agents are to be used to the maximum benefit of patients118.

Acknowledgements
Peerzada Sajad and Konchok Dorjay
Competing Interests
None declared
Author Details
IFFAT HASSAN, MD, Professor and Head, Department of Dermatology, STD and Leprosy, Govt.Medical College Srinagar (University of Kashmir), J & K, India; SAMIA ALEEM, MBBS, Resident, Department of Dermatology, STD and Leprosy, Govt.Medical College Srinagar (University of Kashmir), J & K, India; GOUSIA SHEIKH, MBBS, Resident, Department of Dermatology, STD and Leprosy, Govt.Medical College Srinagar (University of Kashmir), J & K, India; PARVAIZ ANWAR, MD, Senior Resident, Department of Dermatology, STD and Leprosy, Govt.Medical College Srinagar (University of Kashmir), J & K, India.
CORRESPONDENCE: Professor IFFAT HASSAN, Head of Department of Dermatology, STD and Leprosy, Govt.Medical College Srinagar (University of Kashmir), J & K, India.
Email: hassaniffat@gmail.com

References

  1. Feldmann M, Steinman L. Design of effective immunotherapy for human autoimmunity. Nature 2005; 435: 612-9.
  2. Dogra A, Salonie S. Biologic therapy in psoriasis. Indian J Dermatol Venereol Leprol 2006: 72: 256-65.
  3. Stern DK, Tripp JM, Ho VC, Lebwohi M. The use of systemic immune moderators in dermatology. In: Maclean DI, Maddin WS, editors. Dermatologic clinics. Vol. 23. Elsevier; 2005. p. 275.
  4. Waldmann TA. Immunotherapy: Past, present and future. Nat Med 2003; 9: 269-77.
  5. Kormeili T, Lowe NJ, Yamuchi PS. Psoriasis: Immunopathogenesis and evolving immunomodulators and systemic therapies; U.S experiences. Br J Dermatol 2004; 151: 3-15.
  6. Antoni CE, Kruegere GG, de Vlam K, Birbara C, Beutler A, Guzzo C, et al. Infiliximab improves signs and symtoms of psoriatic arthritis the IMPACT 2 trial.Ann Rheum Dis 2005; 64: 1150-7.
  7. Gottlieb AB, Evans R, Li S, Dooley LT, Guzzo CA, Baker D, et al . Infliximab induction therapy for patients with severe plaque type psoriasis: A randomized, double-blind, placebo controlled trial. J Am Acad Dermatol 2004; 51: 534-42.
  8. Winterfield L, Menter A. Psoriasis and its treatment with infliximab-mediated tumour necrosis factor a blockade. Dermatol Clin 2004; 22: 437-44.
  9. Chaudhari U, Romano P, Mulcahy LD, Dooley LT, Baker DG, Gottlieb AB. Efficacy and safety of infliximab montherapy for plaque type psoriasis: A randomized trial. Lancet 2001; 35: 1842-7.
  10. Jacobi A, Antoni C, Manger B, Schuler G, Hertl M. Infliximab in the treatment of moderate to severe atopic dermatitis. J Am Acad Dermatol. 2005; 2: 522-6.
  11. Giamarellos-Bourboulis EJ, Pelekanou E, Antonopoulou A, Petropoulou H, Baziaka F, Karagianni V, et al. An open-label phase II study of the safety and efficacy of etanercept for the therapy of hidradenitis suppurativa. Br J Dermatol 2008; 158: 567-72.
  12. Scheinfeld N. Adalimumab (HUMIRA): A review. J Drugs Dermatol 2003; 2: 375-7.
  13. Gordon KB, Bonish BK, Patel T, Leonardi CL, Nickloff BJ. The tumour necrosis factor-alpha inhibitor adalimumab rapidly reverses the decrease in epidermal Langerhans cell density in psoriatic plaques. Br J Dermatol 2005; 153: 945-53.
  14. Mease PJ, Gladman DD, Ritchlin CT, Ruderman EM, Steinfeld SD, Choy EH, et al. Adalimumab for the treatment of patients with moderately to severely active psoriatic arthritis: Results of a double-blind, randomized, placebo-controlled trial. Arthritis Rheum 2005; 52: 3279-89.
  15. Blanco R, Martínez-Taboada VM, Villa I, González-Vela MC, Fernández-Llaca H, Agudo M, et al. Long-term Successful Adalimumab Therapy in Severe Hidradenitis Suppurativa. Arch Dermatol 2009; 145: 580-84.
  16. Moul DK, Korman NJ. Severe hidradenitis suppurativa treated with adalimumab. Arch Dermatol 2006; 142: 1110-2.
  17. Scheinfeld N. Treatment of coincident seronegative arthritis and hidradenitis suppurativa with adalimumab. J Am Acad Dermatol 2006; 55: 163-4.
  18. Owen CM, Harrison PV. Successful treatment of severe psoriasis with basilixima, an interleukin-2 receptor monoclonal antibody. Clin Exp Dermatol 2000; 25: 195-7.
  19. Salim A, Emerson RM, Dalziel KL. Successful treatment of severe generalized pustular psoriasis with basiliximab (interleukin-2 receptor blocker). Br J Dermatol 2000; 143: 1121-2.
  20. Wohlrab J, Fischer M, Taube KM, Marsch WC. Treatment of recalcitrant psoriasis with daclizumab. Br J Dermatol 2001; 144: 209-10.
  21. Dichmann S, Mrowietz U, Schopf E, Norgauer J. Humanized monoclonal anti-CD25 antibody as a novel therapeutic option in HIV-associated psoriatic erythroderma. J Am Acad Dermatol 2002; 47: 635-6.
  22. Krueger JG, Walters IB, Miyazawa M, Gilleaudeau P, Hakimi J, Light S, et al. Successful in vivo blockade of CD25 (high-affinity interleukin 2 receptor) on T cells by administration of humanized anti-Tac antibody to patients with psoriasis. J Am Acad Dermatol 2000; 43: 448-58.
  23. Langley R, Roenigk HH, McCall C, Stricklin G, Dingivan C. Phase I results of intravenous MEDI-507, an anti-T-cell monoclonal antibody, for the treatment of psoriasis. J Invest Dermatol 2001; 117: 817.
  24. Leonardi CL. Current concepts and review of efalizumab in the treatment of psoriasis. Dermatol Clin 2004; 22: 427-35.
  25. Bohm M, Luger T. Lichen planus responding to efalizumab. J Am Acad Dermatol 2007; 56: 92-3.
  26. Leandro M, Edwards J, Cambridge G. Clinical outcome in 22 patients with rheumatoid arthritis treated with B lymphocyte depletion. Ann Rheum Dis 2002; 61: 883-8.
  27. Anolik JH, Barnard J, Cappione A, Pugh-Bernard AE, Felgar RE, Looney RJ,  et al. Rituximab improves peripheral B cell abnormalities in human systemic lupus erythematous. Arthritis Rheum 2004; 50: 3580-90.
  28. Ahmed A, Spiegelman Z, Cavacini L, Posner M. Treatment of pemphigus vulgaris with rituximab and intravenous immune globulin. N Engl J Med 2006; 355: 1772-9.
  29. Faurschou A, Gniadecki R. Two courses of rituximab for recalcitrant pemphigus vulgaris. Int J Dermatol 2008; 47: 292-4.
  30. Schmidt E, Brockerm E, Goebeler M. Rituximab in treatment-resistant autoimmune blistering skin disorders. Clin Rev Allergy Immunol 2008; 34: 56-64.
  31. Mitra RS, Judge TA, Nestle FO, Turka LA, Nickoloff BJ. Psoriatic skin-derived dendritic cell function is inhibited by exogenous IL-10: differential modulation of B7-1 (CD80) and B7-2 (CD86) expression. J Immunol 1995; 154: 2668-77.
  32. Gottlieb A, Abdulghani A, Totoritis M, Lizambri R, Shuey S, Romano P, et al. Results of a single-dose, dose-escalating trial of an anti-B7 monoclonal antibody (IDEC-114) in patients with psoriasis. J Invest Dermatol 2000; 114: 840.
  33. Gottlieb AB, Kang S, Linden KG, Lebwohl M, Menter A, Abdulghani AA, et al. Results of a multiple-dose, multiple schedule trial of an anti-CD80 monoclonal antibody (IDEC-114) in patients with psoriasis. Poster presented at: annual meeting of the American Academy of Dermatology; March 2, 2001; Washington, DC.
  34. Krueger GG, Langley RG, Leonardi C.A human interleukin-12/23 monoclonal antibody for the treatment of psoriasis. N Engl J Med 2007; 356: 580-92.
  35. Leonardi CL, Kimball AB, Papp KA, Yeilding N, Guzzo C, Wang Y, et al. Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 76-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX). Lancet 2008; 371: 1665-74.
  36. Papp KA, Langley RG, Lebwohl M, Krueger GG, Szapary P, Yeilding N, et al . Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 52-week results from a randomised, double-blind, placebo- controlled trial (PHOENIX). Lancet 2008; 371: 1675-84.
  37. Melmed GY, Targan SR, Yasothan U. Certolizumab pegol. Nat Rev Drug Discov 2008; 7: 641-2.
  38. Hutas G. Golimumab: A fully human monoclonal antibody against TNF alpha. Curr Opin Mol Ther 2008; 10: 393-406.
  39. Rao PE, Kroon DJ. Orthoclone OKT3: Chemical mechanisms and functional effects of degradation of a therapeutic monoclonal antibody. Pharm Biotechnol 1993; 5: 135-58.
  40. Bachelez H, Flageul B, Dubertret L, Fraitag S, Grossman R, Brousse N, et al. Treatment of recalcitrant plaque psoriasis with a humanized non-depleting antibody to CD4. J Autoimmun 1998; 11: 53-62.
  41. Gottlieb AB, Lebwohl M, Shirin S, Sherr A, Gilleaudeau P, Singer G, et al . Anti-CD4 monoclonal antibody treatment of moderate to severe psoriasis vulgaris: Results of a pilot, multicenter, multiple-dose, placebo-controlled study. J Am Acad Dermatol 2000; 43: 595-604.
  42. Barker JN, Jones ML, Mitra RS, et al. Modulation of keratinocyte-derived interleukin-8 which is chemotactic for neutrophils and T lymphocytes. Am J Pathol 1991; 139: 869-76.
  43. Biasi D, Carletto A, Caramaschi P, Bellavite P, Maleknia T, Scambi C, et al. Neutrophil functions and IL-8 in psoriaticarthritis and in cutaneous psoriasis. Inflammation 1998; 22: 533-43.
  44. Krueger GC, Lohner M, Roskos L, Hwang CC, Bell G, Schwab G. Clinical trials results: a fully human anti-IL-8 antibody in patients with moderate to severe psoriasis. Poster presented at: annual meeting of the American Academy of Dermatology; March 2, 2001; Washington, DC.
  45. Lohner M, Krueger J, Gottlieb A, Beutner K, Levy R, Wiesenhutter C, et al. Clinical trials of a fully human anti-IL-8 antibody for the treatment of psoriasis. Br J Dermatol 1999; 141: 989.
  46. Mankad VS. Omalizumab: Other indications and unanswered questions. Clin Rev Allergy Immunol 2005; 29: 17-30.
  47. Spector SL, Tan RA. Effect of omalizumab on patients with chronic urticaria. Ann Allergy Asthma Immunol 2007; 99: 190-3.
  48. Boyce JA. Successful treatment of cold-induced urticaria/anaphylaxis with anti-IgE J Allergy Clin Immunol 2006; 117: 1415-8.
  49. Forman SB, Garrett AB. Success of omalizumab as monotherapy in adult atopic dermatitis: Case report and discussion of the high-affinity immunoglobulin E receptor, Fcepsilon RI. Cutis 2007; 80: 38-40.
  50. Oldhoff JM, Darsow U, Werfel T, Katzer K, Wulf A, Laifaoui J, et al. Anti il-5 recombinant humanised monoclonal antibody for the treatment of atopic dermatitis. Allergy 2005; 60: 693-6.
  51. Simon D, Brathen LR, Simon HU. Anti-interleukin-5 antibody therapy in eosinophilic disorders. Pathabiol 2005; 72: 287-92.
  52. Prashant S, Dennis PW, Kenneth BG. Biologic Therapy for Psoriasis, The New Therapeutic Frontier. Arch Dermatol 2002; 138: 657-63.
  53. Krueger JG. The immunologic basis for the treatment of psoriasis with new biologic agents. J Am Acad Dermatol 2002; 46: 1-23.
  54. Krueger GG. Current concepts and review of alefacept in the treatment of psoriasis. Dermatol Clin 2004; 22: 407-26.
  55. Krueger GG, Papp KA, Stough DB, Loven KH, Gulliver WP, Ellis CN. Alefacept Clinical Study Group. A randomized, double-blind, placebo-controlled phase III study evaluating efficacy and tolerability of 2 courses of alefacept in patients with chronic plaque psoriasis. J Am Acad Dermatol 2002; 47: 821-33.
  56. Heffernan MP, Hurley MY, Martin KS, Smith DI, Anadkat MJ. Alefacept for alopecia areata. Arch Dermatol 2005; 141: 1513-6.
  57. Bui K, Polisetty S, Gilchrist H, Jackson SM, Frederic J. Successful treatment of alopecia universalis with alefacept: a case report and review of the literature. Cutis 2008; 81: 431-4.
  58. Foss C, Clark A, Inabinet R, Camacho F, Jorizzo J. An open label pilot study of alefacept for the treatment of pyoderma gangrenosum. J Eur Acad Derm Venerol 2008; 22: 943-9.
  59. Richardson S, Getfand J. Immunobiologicals, cytokines and growth factors in dermatology. In: Wolff K, Goldsmith LA, Katz SI, Gilchrest BA, Paller AS, Leffell DJ, editors. Fitzpatrick's dermatology in general medicine. 7 th ed. McGraw - Hill; 2008. p. 2223-31.
  60. Moul DK, Routhouska SB, Robinson MR, Korman NJ. Alefacept for moderate to severe atopic dermatitis: A pilot study in adults. J Am Acad Dermatol 2008; 58: 984-9.
  61. Green WH, Leicht SS, Youngberg GA. Patch-stage mycosis fungoides in remission after therapy with alefacept. J Am Acad Dermatol 2008; 58: 110-2.
  62. Chang AL, Badger J, Rehmus W. Alefacept for erosive lichen planus: A case series. J Drugs Dermatol 2008; 7: 379-83.
  63. Hurd DS, Johnston C, Bevins A. A case report of Hailey-Hailey disease treated with alefacept (Amevive). Br J Dermatol 2008; 158: 399-401.
  64. Bagel J, Garland WT, Breneman D, Holick M, Littlejohn TW, Crosby D, et al . Administration of DAB389IL-2 to patients with recalcitrant psoriasis: A double blind, phase II multicenter trial. J Am Acad Dermatol 1998; 3: 938-44.
  65. Moreland L, Bate G, Kirkpatrick P. Abatacept. Nat Rev Drug Discov 2006; 5: 185-6.
  66. Vincenti F, Kirk AD. What's next in the pipeline. Am J Transplant 2008; 8: 1972-81.
  67. Mease PJ, Goffe BS, Metz J, Vander SA, Finck B, Burge DJ. Etanercept in treatment of psoriatic arthritis and psoriasis: A randomized trial. Lancet 2000; 356: 385-90.
  68. Gottlieb AB, Matheson RT, Lowe N, Krueger GG, Kang S, Goffe BS, et al . A randomized trial of etanercept as monotherapy for psoriasis. Arch Dermatol 2003; 139: 1627-32.
  69. Leonardi CL, Powers JL, Matheson RT, Goffe BS, Zitnik R, Wang A, et al . Etanercept as monotherapy in patients with psoriasis. N Engl J Med 2003; 349: 2014-22.
  70. Papp KA, Tyring S, Lahfa M, Prinz J, Griffiths CE, Nakanishi AM, et al . A global Phase III randomized control trial of etanercept in psoriasis: Safety, efficacy and effect of dose reduction. Br J Dermatol 2005; 152: 1304-12.
  71. Goldsmith DR, Wagstaff AJ. Etanercept: A review of its use in the management of plaque psoriasis and psoriatic arthritis. Am J Clin Dermatol 2005; 6: 121-36.
  72. Yamuchi PS, Gindi V, Lowe NJ. The treatment of psoriasis and psoriatic arthritis with etanercept: Practical considerations on monotherapy, combination therapy and safety. Dermatol Clin 2004; 22: 449-59.
  73. Nikas SN, Drosos AA. Onercept. Serono. Curr Opin Investig Drugs 2003; 4: 1369-76.
  74. Holman DM, Kallaji AN. Cytokines in dermatology. J Drug Dermatol 2006; 5: 520-4.
  75. Oppenheim JJ. Cytokines: Past, present and future. Int J Haematol 2001; 74: 3.
  76. Trefzer U, Hofmann M, Sterry W, Asadullah K. Cytokine and anticytokine therapy in dermatology. Expert Opin Biol Ther 2003; 3: 733-43.
  77. Thiel DJ, le Du MH, Walter RL, D'Arcy A, Chène C, Fountoulakis M, et al . Observation of an unexpected third receptor molecule in the crystal structure of human interferon-receptor complex. Structure 2000; 8: 927-36.
  78. Cac NN, Ballas ZK. Recalcitrant warts, associated with natural killer cell dysfunction, treated with systemic IFN-alpha. J Allergy Clin Immunol 2006; 118: 526-8.
  79. Panja SK, Chowdhury A, Banerjee PK, Coondoo A, Nandi S, Saha NC, et al . Interferon Alpha-2b in genital warts. Indian J Dermatol 1991; 36: 5.
  80. Rook AH, Kuzel TM, Olsen EA. Cytokine therapy of cutaneous T-cell lymphoma:         Interferons, interleukin-12, and interleukin-2. Hematol Oncol Clin North Am 2003; 17: 1435-8.
  81. Potthoff A, Brockmeyer NH. HIV-associated Kaposi sarcoma: Pathogenesis and therapy. J Dtsch Dermatol Ges 2007; 5: 1091-4.
  82. Kalani AD, Jack A, Montenegro G, Degliuomini J, Wallack MK. Immunotherapy as an adjuvant therapy in the management of advanced, surgically resected melanoma. G Ital Dermatol Venereol 2008; 143: 59-70.
  83. Tucker SB, Polasek JW, Perri AJ, Goldsmith EA. Long-term follow-up of basal cell carcinomas treated with perilesional interferon alfa 2b as monotherapy. J Am Acad Dermatol 2006; 54: 1033-8.
  84. Edwards L, Berman B, Rapini RP, Whiting DA, Tyring S, Greenway HT Jr, et al . Treatment of cutaneous squamous cell carcinomas by intralesional interferon alpha 2b therapy. Arch Dermatol 1992; 128: 1486-9.
  85. Edwards L, Levine N, Widener M. Effect of intralesional alpha 2 interferon on Actinic keratosis. Arch Dermatol 1986; 122: 779.
  86. Tsambaos D, Eichelberg D, Goos M. Behçet's syndrome: Treatment with recombinant leucocyte alpha-interferon. Arch Dermatol Res 1986; 278: 335-6.
  87. Kim HJ, Colombo M, Frieden IJ. Ulcerated haemangiomas: Clinical characteristics and response to therapy. J Am Acad Dermatol 2001; 44: 962-72.
  88. Al-Attar A, Mess S, Thomassen JM., Kauffman CL, Davison SP. Keloid pathogenesis and treatment. Plast Reconstr Surg 2006; 117: 286-300.
  89. Gross G, Ikenberg H, Roussaki A, Drees N, Schöpf E. Systemic therapy of condylomata acuminata with recombinant alpha 2a: Low dose superior to the high dose regimen. Chemotherpay 1986; 32: 537-41.
  90. Mitchell MS, Abrams J, Thompson JA, Kashani-Sabet M, DeConti RC, Hwu WJ, et al . Randomized trial of an allogeneic melanoma lysate vaccine with low-dose interferon Alfa-2b compared with high-dose interferon Alfa-2b for Resected stage III cutaneous melanoma. J Clin Oncol 2007; 25: 2078-85.
  91. Bolinger A, Taeubel MA. Recombinant interferon gamma for treatment of chronic granulomatous disease and other disorders. Clin Pharm 1992; 11; 834-50.
  92. Hanifin JM, Schneider LC, Leung DY, Ellis CN, Jaffe HS, Izu AE, et al . Recombinant interferon gamma therapy for Atopic Dermatitis. J Am Acad Dermatol 1993; 28: 189-97.
  93. Pierer M, Baerwald C. Biological therapy for the treatment of rheumatic diseases. Internist (Berl) 2008; 49: 938-46.
  94. Frischmeyer-Guerrerio PA, Rachamalla R, Saini SS. Remission of Schnitzler syndrome after treatment with anakinra. Ann Allergy Asthma Immunol 2008; 100: 617-9.
  95. Ross JB, Finlayson LA, Klotz PJ, Langley RG, Gaudet R, Thompson K, et al . Use of anakinra (Kineret) in the treatment of familial cold autoinflammatory syndrome with a 16-month follow-up. J Cutan Med Surg 2008; 12: 8-16.
  96. Peddle L, Butt C, Snelgrove T. Interleukin (IL) 1alpha, IL1beta, IL receptor antagonist, and IL10 polymorphisms in psoriatic arthritis. Ann Rheum Dis 2005; 64: 1093-4.
  97. Marzec M, Halasa K, Kasprzycka M, Wysocka M, Liu X, Tobias JW, et al . Differential effects of interleukin-2 and interleukin-15 versus interleukin-21 on CD4 + cutaneous T-cell lymphoma cells. Cancer Res 2008; 68: 1083-91.
  98. Atkins MB, Lotze MT, Dutcher JP, Fisher RI, Weiss G, Margolin K, et al . High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: Analysis of 270 patients treated between 1985 and 1993. J Clin Oncol 1999; 17: 2105-16.
  99. Ghoreschi K, Thomas P, Breit S, Dugas M, Mailhammer R, van Eden W, et al . Interleukin-4 therapy of psoriasis induces Th2 responses and improves human autoimmune disease. Nat Med 2003; 9: 40-6.
  100. Trepicchio WL, Ozawa M, Walters IB, Kikuchi T, Gilleaudeau P, Bliss JL, et al . Interleukin-11 therapy selectively down regulates type I cytokine proinflammatory pathways in psoriasis lesions. J Clin Invest 1999; 104: 1527-37.
  101. Conti L, Gessani S. GM-CSF in the generation of dendritic cells from human blood monocyte precursors: Recent advances. Immunobiology 2008; 213: 859-70.
  102. Chaperot L, Chokri M, Jacob MC, Drillat P, Garban F, Egelhofer H, et al . Differentiation of antigen-presenting cells (dendritic cells and macrophages) for therapeutic application in patients with lymphoma. Leukemia 2000; 14: 1667-77.
  103. Meier K, Nanney LB. Emerging new drugs for wound repair. Expert Opin Emerg Drugs 2006; 11: 23-37.
  104. Alikhan MA, Carter G, Mehta P. Topical GM-CSF hastens healing of leg ulcers in sickle cell disease. Am J Hematol 2004; 76: 192.
  105. Elias EG, Zapas JL, McCarron EC, Beam SL, Hasskamp JH, Culpepper WJ. Sequential administration of GM-CSF (Sargramostim) and IL-2 ± autologous vaccine as adjuvant therapy in cutaneous melanoma: An interim report of a phase II clinical trial. Cancer Biother Radiopharm 2008; 23: 285-91.
  106. Bouwhuis SA, Markovic SN, McEvoy MT. Extracorporeal photopheresis and adjuvant aerosolized granulocyte-macrophage colony-stimulating factor for Sézary syndrome. Mayo Clin Proc 2002; 77: 197-200.
  107. Pinkas H, Fisch B, Rozansky G, Felz C, Kessler-Icekson G, Krissi H, et al . Platelet-derived growth factors (PDGF-A and -B) and their receptors in human fetal and adult ovaries. Mol Hum Reprod 2008; 14: 199-206.
  108. Embil JM, Papp K, Sibbald G. Recombinant human platelet-derived growth factor-BB (becaplermin) for healing chronic lower extremity diabetic ulcers: An open-label clinical evaluation of efficacy. Wound Repair Regen 2000; 8: 162-8.
  109. Asadullah K, Döcke WD, Ebeling M, Friedrich M, Belbe G, Audring H, et al. Interleukin 10 treatment of psoriasis: clinical results of a phase II trial. Arch Dermatol 1999; 135: 187-192.
  110. Smith CH, Anstey AV, Barker JN, Burden AD, Chalmers RJ, Chandler D, et al. British association of Dermatologists guidelines for use of biological interventions in psoriasis 2005. Br J Dermatol 2005; 153: 486-97.
  111. Sterry W, Barker J, Boehncke WH, Bos JD, Chimenti S, Christophers E, et al. Biological therapies in the systemic management of psoriasis: International Consensus Conference. Br J Dermatol 2004; 151: 3-17.
  112. Koo J, Khera P. Update on the mechanisms and efficacy of biological therapies for psoriasis. J Dermatol Sci 2005; 38: 75-87.
  113. Asadullah K, Sterry W, Ebeling M, Friedrich M, Leupold M, Jasulaitis D, et al. Clinical and immunological effects of IL-10 therapy in psoriasis. Br J Dermatol 1999; 141: 989.
  114. Sukal SA, Nadiminti L, Granstein RD. Etanercept and demyelinating disease in a patient with psoriasis. J Am Acad Dermatol 2006; 54: 160-4.
  115. Scheinfeld N. Adalimumab: A review of side effects. Exp Opin Drug Saf 2005; 4: 637-41.
  116. Thappa DM, Laxmisha C. Immunomodulators in the treatment of Psoriasis. Indian J Dermatol Venereol Leprol 2004; 70: 1-9.
  117. Lebowhl M, Clark LN. Recent advances in medical dermatology. Int J Dermatol 2007; 1: 197-202.
  118. Smith CH, Anstey AV, Barker JN, Burden AD, Chalmers RJ, Chandler DA, et al. British association of dermatologist guidelines for biological intervention for psoriasis 2009.Br J Dermatol 2009; 161: 987-1019.


Creative Commons Licence
The above article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.


share