Alopecia Areata

Number: 0423

Table Of Contents

Policy
Applicable CPT / HCPCS / ICD-10 Codes
Background
References


Policy

Scope of Policy

This Clinical Policy Bulletin addresses alopecia areata.

  1. Medical Necessity

    Aetna considers the following treatments medically necessary for alopecia areata when criteria are met:

    1. For mild alopecia areata (less than 50 % loss of scalp hair):

      1. Anthralin (topical)
      2. Glucocorticoid (topical, intralesional);
    2. For extensive alopecia areata (greater than 50 % loss of scalp hair):

      1. Anthralin (topical)
      2. Glucocorticoid (oral, topical, intralesional)
      3. Psoralen (oral or topical) photochemotherapy (PUVA) (see CPB 0205 - Phototherapy and Photochemotherapy (PUVA) for Skin Conditions);
    3. Topical immunotherapy (i.e., diphenylcyclopropenone [DPCP/DCP], squaric acid dibutyl ester [SADBE]) for extensive alopecia areata (greater than 50 % loss of scalp hair) when conventional therapies have failed.

  2. Experimental and Investigational

    Aetna considers the following therapies / tests experimental and investigational for alopecia areata (AA) because the effectiveness of these approaches has not been established (not an all-inclusive list):

    1. Therapies for AA:

      1. Acupuncture
      2. Adalimumab
      3. Aromatherapy
      4. Azathioprine
      5. Botulinum toxin (including intradermal)
      6. Brevilin A (systemic and topical)
      7. Capsaicin
      8. Carboxytherapy (transcutaneous infusion of carbon dioxide into the affected site)
      9. Compound glycyrrhizin
      10. Cryotherapy
      11. Cyclosporine
      12. Dinitrochlorobenzene (DNCB)
      13. Etanercept
      14. Excimer laser
      15. Extracorporeal photopheresis
      16. Fractional carbon dioxide laser
      17. Fractional photothermolysis
      18. Hydroxychloroquine
      19. Hypnosis
      20. Infliximab
      21. Inosiplex
      22. Intralesional Candida antigen immunotherapy
      23. Laser and light therapy combined with topical minoxidil
      24. Low-dose naltrexone
      25. Low-level laser therapy
      26. Mesenchymal stem cells
      27. Methotrexate
      28. Mindfulness psychotherapy
      29. Narrow-band ultraviolet B phototherapy
      30. Phosphatidylcholine
      31. Photodynamic therapy
      32. Platelet-rich fibrin
      33. Platelet-rich plasma
      34. Prostaglandins (e.g., bimatoprost and latanoprost)
      35. Psychological interventions
      36. Recombinant interleukin 2 (e.g., aldesleukin [Proleukin])
      37. Topical calcipotriol
      38. Topical garlic
      39. Topical triiodothyronine
      40. Total glucosides of peony (paeony)
      41. Ustekinumab
      42. Zinc supplementation;
    2. Tests for AA:

      1. CTLA4
      2. FAS
      3. FASL
      4. Glutathione peroxidase (GPx1 Pro 197 Leu)
      5. HLA-DRB1
      6. IL2RA
      7. Interleukin-10 (IL-10), IL-12, IL-17, IL-18, IL-23, and IL-35
      8. Manganese superoxide dismutase (MnSODAla-9Val)
      9. Non-receptor type 22 (PTPN22) and transporter 1, ATP-binding cassette sub-family B (MDR/TAP) gene (TAP1) gene polymorphisms 
      10. Protein tyrosine phosphatase
      11. Evaluation of serum trace elements (e.g., copper, magnesium, selenium, and zinc) level for the diagnosis of AA
      12. Measurement of inflammatory biomarkers (red cell distribution width [RDW], mean platelet volume [MPV], plateletcrit [PCT], the ratio of neutrophils to lymphocytes [NLR], and the ratio of platelets to lymphocytes [PLR]).
  3. Related Policies

    For baricitinib (Olumiant), please refer to pharmacy benefit plans.

    See also:

    1. CPB 0205 - Phototherapy and Photochemotherapy (PUVA) for Skin Conditions.

Table:

CPT Codes / HCPCS Codes / ICD-10 Codes

Code Code Description

CPT codes covered if selection criteria are met:

11900 Injection, intralesional; up to and including seven lesions
11901     more than seven lesions
96912 Photochemotherapy; psoralens and ultraviolet A (PUVA)

CPT codes not covered for indications listed in the CPB:

Interleukin-12, interleukin -17, Interleukin-18 gene polymorphisms testing, ATP-binding cassette sub-family B (MDR/TAP) gene (TAP1) gene polymorphisms, protein tyrosine phosphatase, non-receptor type 22 (PTPN22), transporter 1, Carboxytherapy, Cryotherapy, fractional carbon dioxide laser, FAS, FASL, CTLA4 and IL2RA gene polymorphisms testing, Measurements of serum levels of Interleukins 10 (IL-10), Il-12, Il-17 and Il-35, Platelet-rich fibrin - no specific code
0232T Injection(s), platelet rich plasma, any site, including image guidance, harvesting and preparation when performed
0481T Injection(s), autologous white blood cell concentrate (autologous protein solution), any site, including image guidance, harvesting and preparation, when performed
0552T Low-level laser therapy, dynamic photonic and dynamic thermokinetic energies, provided by a physician or other qualified health care professional
36522 Photopheresis, extracorporeal
38205 Blood derived hematopoietic progenitor cell harvesting for transplantation, per collection allogeneic
38206 Blood derived hematopoietic progenitor cell harvesting for transplantation, autologous
81382 HLA Class II typing, high resolution (ie, alleles or allele groups); one locus (eg, HLA-DRB1, -DRB3/4/5, -DQB1, -DQA1, -DPB1, or -DPA1), each [not covered for HLA-DRB1 gene polymorphisms testing]
82495 Chromium
82525 Copper
83540 Iron
83735 Magnesium
84255 Selenium
84630 Serum zinc
85049 Blood count; platelet, automated
86003 Allergen specific IgE; quantitative or semiquantitative, each allergen
90853 Group psychotherapy (other than of a multiple-family group) [mindfulness psychotherapy]
90880 Hypnotherapy
96156 Health behavior assessment, or re-assessment (ie, health-focused clinical interview, behavioral observations, clinical decision making)
96158 Health behavior intervention, individual, face-to-face; initial 30 minutes
96159      each additional 15 minutes (List separately in addition to code for primary service)
96164 Health behavior intervention, group (2 or more patients), face-to-face; initial 30 minutes
96165      each additional 15 minutes (List separately in addition to code for primary service)
96167 Health behavior intervention, family (with the patient present), face-to-face; initial 30 minutes
96168      each additional 15 minutes (List separately in addition to code for primary service)
96170 Health behavior intervention, family (without the patient present), face-to-face; initial 30 minutes
96171      each additional 15 minutes (List separately in addition to code for primary service).
96567 Photodynamic therapy by external application of light to destroy pre-malignant and/or malignant lesions of the skin and adjacent mucosa (e.g., lip) by activation of photosensitive drug(s) each phototherapy exposure session
96573 Photodynamic therapy by external application of light to destroy premalignant lesions of the skin and adjacent mucosa with application and illumination/activation of photosensitizing drug(s) provided by a physician or other qualified health care professional, per day
96900 Actinotherapy (ultraviolet light)
96910 Photochemotherapy; tar and ultraviolet B (Goeckerman treatment) or petrolatum and ultraviolet B
97810 - 97814 Acupuncture

CPT codes related to the CPB:

96372 Therapeutic, prophylactic, or diagnostic injection (specify substance or drug), subcutaneous or intramuscular

HCPCS codes not covered for indications listed in the CPB:

Topical garlic, compound glycyrrhizin, Brevilin A, Intralesional candida antigen immunotherapy - no specific code
J0135 Injection, adalimumab, 20 mg
J0585 Injection, onabotulinumtoxinA, 1 unit
J0586 Injection, AbobotulinumtoxinA, 5 units
J0587 Injection, rimabotulinumtoxinB, 100 units
J1438 Injection, etanercept, 25 mg (code may be used for Medicare when drug administered under the direct supervision of a physician, not for use when drug is self administered)
J1745 Injection, infliximab, 10 mg
J2315 Injection, naltrexone, depot form, 1 mg
J7308 Aminolevulinic acid HCL for topical administration, 20%, single unit dosage form (354 mg)
J7335 Capsaicin 8% patch, per 10 square centimeters
J7336 Capsaicin 8% patch, per square centimeter
J7500 Azathioprine, oral 50 mg
J7501 Azathioprine, parenteral, 100 mg
J7502 Cyclosporine, oral 100 mg
J7515 Cyclosporine, oral 25 mg
J7516 Cyclosporine, parenteral 250 mg
J8610 Methotrexate, oral 2.5 mg
J9015 Injection, aldesleukin, per single use vial
J9250 Methotrexate sodium, 5 mg
J9255 Injection, methotrexate (accord) not therapeutically equivalent to j9250 or j9260, 50 mg
J9260 Methotrexate sodium, 50 mg
P9020 Platelet rich plasma, each unit
Q5131 Injection, adalimumab-aacf (idacio), biosimilar, 20 mg
Q5132 Injection, adalimumab-afzb (abrilada), biosimilar, 10 mg
S0139 Minoxidil, 10 mg
S8948 Application of a modality (requiring constant provider attendance) to one or more areas; low-level laser; each 15 minutes

HCPCS codes related to the CPB:

P9022 Red blood cells, washed, each unit

ICD-10 codes covered if selection criteria are met:

L63.0 - L63.9 Alopecia areata

ICD-10 codes not covered for indications listed in the CPB:

L64.9 Androgenic alopecia, unspecified

Background

Alopecia areata (AA) is a disease characterized by hair cycle dysfunction and the presence of peribulbar and perifollicular mononuclear cell infiltrates.  The diagnosis of this condition is made by observation.  The majority of patients is under 40 years old and report the rapid onset of one or several defined, usually round, 1 to 4 cm areas of scalp hair loss.  A common feature is the presence of “exclamation-mark” hairs that may be present at the margins of the bald patch.  “Exclamation-mark” hairs are broken, short hairs that taper proximally.  Some patients with alopecia areata also exhibit nail pitting.  The disease may affect any hair-bearing area, but most commonly affects the scalp, eyebrows, eyelashes, and beard.  Hair loss may be patchy or extensive.  In extreme cases, the disease may result in total loss of scalp hair (alopecia totalis) or scalp and body hair (alopecia universalis).

Although the etiology of alopecia areata is unknown, most evidence supports the hypothesis that the disease is immunologically mediated.  Circulating autoantibodies and follicular deposits of C3 and IgG have been reported.  Alopecia areata usually occurs as an isolated condition, but may occur in conjunction with pernicious anemia, thyroid disease, ulcerative colitis, Addison's disease, vitiligo, lupus erythematosus, and Down syndrome.

Treatment success depends on the age of onset of the disease and the extent of hair loss.  The prognosis tends to be worse in more extensive cases (alopecia totalis or universalis), or when alopecia areata begins in early childhood.  In all cases, hair regrowth may occur spontaneously without treatment, even after months or years.  In both mild and extensive cases of alopecia areata, topical corticosteroids of medium to very high potency are used.

The most common treatment for mild cases of alopecia areata (involving less than 50 % loss of scalp hair) is direct intradermal injection of corticosteroids (e.g., cortisone or triamcinolone acetonide) into patches of hair loss.  Multiple injections are administered monthly to the skin in and around the bare patches; an average of 4 to 6 monthly injections are usually required for significant improvement.  The prognosis for total permanent regrowth in cases with limited involvement is excellent.  Topical glucocorticoid therapy may be used alone or in combination with other therapies, such as anthralin or injected glucocorticoids.

Anthralin is a synthetic, tar-like substance that has been widely used for psoriasis.  Anthralin's effectiveness in inducing hair regrowth may be due to a non-specific immunomodulating effect.  It is potentially irritating and may cause redness, itching, and scaling; therefore, it is often applied and then removed 20 to 60 mins later (short-contact therapy).

Therapy for extensive alopecia areata (involving more than 50 % loss of scalp hair) may be prolonged and difficult.  Systemic corticosteroids are seldom used due to their adverse effects; however, they may be required depending on the severity of the condition and the adequacy of the response to topical therapy.  Treatments usually need to be continued until remission of the disease occurs; however, if there is no significant response after 6 months of treatment, oral corticosteroids are unlikely to be effective.

Psoralen photochemotherapy (psoralen and ultraviolet light A or PUVA) is another immunosuppressant treatment that is used for alopecia areata.  The psoralen is administered topically or orally and is followed by 1 or 2 hours with UVA (ultraviolet A); however, between 40 and 80 treatments may be required before hair regrowth occurs.  The need for long-term therapy, along with concern about increased risk of photodamage/photoaging and skin cancer, make PUVA therapy less than satisfactory.  In addition, its effectiveness has been questioned in the literature.  Healy and Rogers (1993) reported on the results of 102 alopecia areata patients treated with PUVA and concluded that the results achieved with PUVA differed little from what would be expected with no treatment.  Furthermore, there is a high relapse rate when PUVA treatment is discontinued.  Despite these limitations, PUVA is still considered standard practice of care by the American Academy of Dermatology.

Topical immunotherapy has been used in Canada and Europe with reported hair growth rates of 40 to 60 % among patients with scalp hair loss of 50 to 99 %.  Topical immunotherapy with DPCP/DCP and SADBE is offered only at a few centers in the United States due to the investigational status of these drugs for alopecia areata.  Initial responses are generally seen after 12 weeks of therapy with cosmetically acceptable results in 24 weeks.  If there is no response by the end of 24 weeks, immunotherapy is discontinued.  Over the long-term, approximately 1/3 of patients eventually stop responding to therapy.

Topical minoxidil (Rogaine) and oral finasteride (Propecia) are indicated for androgenetic alopecia (male-pattern hair loss); they have not been approved by the Food and Drug Administration (FDA) for treatment of alopecia areata.

Tacrolimus ointment is a steroid-free topical immunomodulator developed for the treatment of atopic dermatitis.  By inhibiting T-cell activation and cytokine production, topically applied tacrolimus modulates inflammatory responses in the skin.  Many studies have shown that it is effective and well-tolerated for the treatment of atopic dermatitis.  Moreover, it has been suggested that tacrolimus ointment may be effective treatment for a variety of other inflammatory skin disorders such as alopecia areata.  Price and colleagues (2005) reported their findings on the use of topical tacrolimus for patients with alopecia areata.  These researchers found that 11 patients with alopecia areata affecting 10 to 75 % of the scalp with an average duration 6 years had no terminal hair growth in response to tacrolimus ointment 0.1% applied twice-daily for 24 weeks.

In a review on the diagnosis and treatment of iron deficiency and its potential relationship to hair loss, Trost et al (2006) stated that there is insufficient evidence to recommend universal screening for iron deficiency in patients with hair loss.  In addition, there is insufficient evidence to recommend giving iron supplementation therapy to patients with hair loss and iron deficiency in the absence of iron deficiency anemia.

Willemsen et al (2006) noted that only limited data exist on the role of psychotherapy in alopecia areata.  These investigators sought to document the influence of hypnotherapy on psychological well-being and clinical outcome in patients with alopecia areata.  Hypnosis was used in 28 patients with extensive alopecia areata who were refractory to previous conventional treatments.  It was added as a complementary treatment or used as the only treatment.  In all, 21 patients (9 with alopecia totalis or alopecia universalis and 12 with extensive alopecia areata) were analyzed during a 5-year period.  After treatment, all patients had a significantly lower score for anxiety and depression.  Scalp hair growth of 75 % to 100 % was seen in 12 patients after 3 to 8 sessions of hypnotherapy.  Total growth occurred in 9 of these 12 patients, including 4 patients with alopecia universalis and 2 with ophiasis.  In 5 patients, a significant relapse occurred.  The authors concluded that hypnotherapy may enhance the mental well-being of patients with alopecia areata and it may improve clinical outcome.  However, they noted that this is a preliminary study with a limited number of patients, and a larger randomized controlled trial is need to validate these early findings.

In a Cochrane review on interventions for alopecia areata, Delamere and colleagues (2008) evaluated a range of interventions that included topical photodynamic therapy and topical minoxidil.  Overall, none of the interventions showed significant treatment benefit in terms of hair growth when compared with placebo.

Faghihi and associates (2009) noted that latanoprost is an analog of prostaglandin F(2-alpha) that is used to treat glaucoma.  Increases in eyelash number, thickness, and pigmentation have been reported as latanoprost side effects.  These investigators examined if topical use of this drug can be used as a treatment of alopecia areata of eyebrows and eyelashes or not.  In an experimental study, 26 patients with symmetrical eyelash and eyebrow alopecia areata were treated over 4 months with topical latanoprost for one side and the other side was not treated with any drug.  The results were compared.  Only 1 of the latanoprost-treated cases showed partial hair regrowth on the treated side.  The relationship between hair regrowth and latanoprost application was not statistically significant (p = 1) by Fisher test.  Based on these findings, topical latanoprost is not effective in the treatment of alopecia areata.  The authors stated that more studies with a larger sample size, longer study duration, and higher concentration of medication are needed.

In a 2-year, prospective, non-blinded, non-randomized, controlled study, Coronel-Perez et al (2010) examined the effectiveness of latanoprost in eyelash alopecia areata.  These investigators conducted a survey of 54 subjects with alopecia areata universalis; control group comprised 10 subjects who received injections of 0.5 mg/cm(2) of triamcinolone acetonide (TAC) in their eyebrows and 1 mg/cm(2) of TAC injections in affected scalp.  The treatment group included 44 subjects who received the same treatment as the control group in scalp and eyebrows but they also applied a drop of latanoprost 0.005 % (50 microg/ml) ophthalmic solution in their eyelid margins every night.  Subjects were reviewed every 3 months for 2 years.  A total of 40 subjects finished the study and 4 subjects were lost to follow-up.  In the treatment arm of this study, the course was well-tolerated and uncomplicated.  Both investigators and patients evaluated the regrowth.  The results obtained were: complete regrowth in 17.5 %, moderate regrowth in 27.5 %, slight regrowth in 30 % and without response in 25 %.  Moderate and total regrowth constituted a cosmetically acceptable response.  The therapy was continuous and the response remained without any side effects.  No patients had cosmetically acceptable eyelash regrowth in the control group.  The authors concluded that latanoprost may be an effective drug in the treatment of eyelash alopecia areata because it induces acceptable responses (total and moderate) in 45 % of the patients.  They stated that a formal, blinded, prospective unilateral controlled study will permit further understanding about this promising therapeutic agent for eyelash alopecia areata.

In a review on alopecia areata, Alkhalifah and co-workers (2010) noted that several reports of multiple biologics, including adalimumab, efalizumab, etanercept, and infliximab failed to show improvement in patients with alopecia areata.  Furthermore, these investigators stated that the use of topical calcineurin inhibitors (e.g., pimecrolimus and tacrolimus) in alopecia areata was unsuccessful.  In a single study, bexarotene 1 % gel resulted in a 26 % hair regrowth rate; dermal irritation is a common side effect.  These researchers stated that the effectiveness of bexarotene needs to be confirmed in randomized, placebo-controlled trials.  Capsaicin was previously reported to induce vellus hair regrowth in alopecia areatad.  More recently, a study showed that topical capsaicin and clobetasol 0.05 % are comparable.  Moreover, these investigators stated that these findings should be supported by randomized, placebo-controlled trials before capsaicin use is added to the therapeutic armamentarium of alopecia areata.  Ustekinumab, a fully human monoclonal antibody to the shared p40 subunit of interleukin-12 and interleukin-23, has been shown to be effective in plaque psoriasis, and studies are ongoing to evaluate its long-term safety and effectiveness.  The authors stated that ustekinumab may be tried on patients with alopecia areata in the future.  In addition, these researchers noted that the relation between vitamin D levels and the development of alopecia areata, and whether vitamin D supplementation helps in the treatment of alopecia areata represent an attractive area of research, the results of which may prove that vitamin D is a safe and helpful choice in the treatment of alopecia areata.

In a pilot study, Farshi et al (2010) evaluated the safety and effectiveness of azathioprine as a systemic monotherapy for moderate-to-severe alopecia areata.  A total of 20 patients (14 men [70 %] and 6 women [30 %]) with minimum 6 months history of alopecia areata were included.  The extent of scalp hair regrowth during and after the completion of the 6 months treatment was evaluated by the Severity of Alopecia Tool (the SALT score).  The daily drug intake was calculated as 2 mg/kg of body weight.  Mean duration of current episode of scalp hair loss was 26.4 (26.4 +/- 17) months.  Mean regrowth percentage was 52.3 % (52.3 +/- 38.4). Mean hair loss percentage before treatment was 72.7 % (72.7 +/- 28.3) compared with 33.5 % (33.5 +/- 30.7) after 6 months of azathioprine treatment.  This showed a highly significant statistical difference (paired t-test, confidence interval [CI]: 95 %: 21.5 to 54.1).  Mean hair loss score (S(0)  to S(5)) before treatment was 3.9 (3.9 +/- 1.6) and after 6 months of azathioprine treatment was 1.8 (1.8 +/- 1.3).  Assessment showed significant difference from baseline score (sign test, p < 0.0001).  No significant statistical difference was observed with respect to gender before and after azathioprine treatment.  Treatment with azathioprine as a systemic monotherapy clinically produces relevant improvement in moderate-to-severe alopecia areata.  The authors concluded that generally azathioprine is a low-cost and well-tolerated drug and with controlled studies on larger number of patients, long-term safety and effectiveness of this treatment should be investigated.

Cho and colleagues (2010) examined the safety and efficacy of botulinum toxin type A (BTXA) injections for the treatment of patients with alopecia areata of the scalp.  A total of 7 patients with alopecia areata received 10 U of BTXA intradermal injections on each site 3 times.  Subjects were classified according to the extent of scalp hair loss into Severity of Alopecia Tool subclasses.  Two patients had one patch of alopecia areata; the remaining patients had total or universal type alopecia areata.  One patient dropped out of the study after experiencing spontaneous recovery from her alopecia areata.  One patient reported aggravation of her alopecia areata following BTXA injections.  The remaining patients' alopecia areata did not change after BTXA injections.  The authors concluded that these findings suggested that BTXA injection can not be used as an alternative treatment for recalcitrant alopecia areata.  Nevertheless, future studies concerning the treatment efficacy of BTXA for mild-to-moderate alopecia areata are needed.

Bayramgürler and colleagues (2011) stated that although narrow-band ultraviolet B (NB UVB) phototherapy is a well-established treatment in many dermatosis, there is little evidence of efficacy of this method for alopecia areata (AA) treatment in the literature.  These investigators undertook a retrospective review of the 25 AA patients treated with NB UVB.  Intra-muscular triamcinolone acetonide injections per month were used as concomitant treatment in some patients who did not have any contraindication.  Eight patients (32 %) received monthly intra-muscular corticosteroid injections.  Four (22.2 %) and 2 (20 %) patients achieved excellent response in extensive patchy hair loss patients and entire scalp hair loss patients, respectively.  Four of 6 patients who achieved excellent response also received monthly intra-muscular corticosteroid injections.  When patients receiving systemic corticosteroid injections were compared with patients given only NB UVB with respect to the treatment responses, a statistically significant difference was seen in patients who achieved excellent response.  Narrow-band UVB is not an effective treatment with only 20 % excellent treatment responses in patients with severe AA, most of whom were also treated with systemic corticosteroids.

In a double-blind, randomized pilot clinical trial, Nasiri et al (2012) examined the efficacy of topical triiodothyronine in patients with patchy AA.  A total of 10 patients with patchy AA were treated with triiodothyronine and placebo applied twice-daily to either of 2 bilaterally symmetrical patches for 12 weeks.  The 2 sides were randomly assigned following simple randomization procedure to one of the two treatment groups.  The patients and the investigator were blinded to the content of the tubes.  Hair regrowth was evaluated every 4 weeks.  Blood samples for measurements of complete blood count along with thyroid function (T3, T4 and TSH) and liver function tests were taken at the baseline and at the end of study.  After 12 weeks of treatment, there was no statistically significant difference between the outcome in terms of reduction of the patch size and hair regrowth.  No adverse effects were noted.  The authors concluded that triiodothyronine in the studied dosage and formulation was safe but not more effective than placebo.

Park et al (2013) examined if the combination therapy of cyclosporine and psoralen plus ultraviolet A (PUVA) could be an effective treatment for severe AA.  A total of 41 patients with severe AA were treated with oral cyclosporine and topical PUVA.  Cyclosporine was given at an initial daily dose of 200 mg for adult and 100 mg for children for periods of up to 16 weeks.  Eight-methoxypsoralen (Methoxsalen) was applied topically 20 minutes prior to ultraviolet A (UVA) exposure, and the patients were irradiated with UVA twice-weekly for 16 weeks.  Of the total 41 patients, 2 (7.3 %) patients were lost to follow-up, and 1 (2.4 %) patient discontinued the treatment due to abdominal discomfort.  Six (14.6 %) patients were treated for less than 12 weeks.  Of remaining 32 patients, 3 (9.4 %) showed excellent response, 3 (9.4 %) showed good response, 12 (37.5 %) showed fair response, and 14 (43.7 %) showed poor response.  The authors concluded that although limited by its uncontrolled character, this study showed that the combination therapy with cyclosporine and PUVA may be an additional choice for severe and recalcitrant AA.

Staumont-Salle et al (2012) evaluated the long-term outcomes of patients with AA who were treated with methylprednisolone bolus.  This study included 60 patients treated between 1995 and 2000.  The short-term outcomes were analyzed in 2000.  The long-term assessment of 30 patients was performed in 2010 by phone questionnaire.  Significant hair regrowth was observed in 10/30 patients at 6 months after the bolus treatment.  Half of the plurifocalis AA patients were responders at 6 months, but less than 25 % of alopecia totalis (AT) and alopecia universalis (AU) patients responded.  Long-term outcomes were assessed after a mean duration of 12.3 years; 8/10 initial responders had mild or no disease, and 14/20 initial non-responders had severe AA.  The authors concluded that this study confirmed the low efficiency, both short- and long-term, of this treatment for AT and AU.

Bin Saif et al (2012) examined the safety and effectiveness of oral mega pulse methylprednisolone for patients with severe therapy resistant AA.  Patients with AU, AT, or alopecia ophiasis (AO) were assigned to one of the 3 treatment groups: Group A received oral mega pulse methylprednisolone (MP) for 3 consecutive days once every 2 weeks for 24 weeks; Group B received 2 consecutive daily pulses every 3 weeks; and Group C received 3 consecutive daily pulses every 3 weeks.  Patients who showed regrowth of 75 % or more at 24 or 36 weeks continued their treatment, while intervals were increased gradually.  A total of 42 patients were included in this study, and 52.4 % of them had atopic diathesis, while 35.7 % had autoimmune thyroiditis.  At 36 weeks, 12 (28.6 %) patients had adequate response, 9 (21.4 %) had inadequate response, and 21 (50 %) patients had poor response.  The response rate showed no statistically significant difference between treatment groups.  There were statistically significant differences in age of onset, duration of the disease, and presence of subclinical hypothyroidism between different response groups.  At follow-up: 13 (38.2 %) patients relapsed; 5 (14.7 %) patients developed moderate hair fall; 3 (8.8 %) patients developed mild hair fall; 7 (20.1 %) patients maintained their hair regrowth; and 6 (17.6 %) patients were lost in follow-up.  Treatment was relatively well-tolerated among subjects in groups B and C.  The authors concluded that oral mega pulse MP use in severe forms of AA has relative effectiveness and tolerance; but with high relapse rate.

In a retrospective case-series study, Droitcourt et al (2012) examined the safety and effectiveness of combination of systemic pulse corticosteroids and methotrexate in the treatment of severe AA.  Patients were treated with intravenous 500 mg methylprednisolone per day for 3 consecutive days monthly during 3 months plus methotrexate initiated at the end of the second pulse regimen.  These investigators reviewed all case notes of patients who received this regimen between January 1 2007 and December 1 2010.  A total of 20 patients were treated.  Data on hair regrowth at month 12 were available for all patients; 14 patients were still receiving the treatment on December 1 2010, 2 patients were lost in follow-up, and 4 patients had stopped the treatment.  Of the 14 patients who were still receiving the treatment regimen at month 18, 10 (10/20, 50 %) had total hair regrowth and 4 (4/20, 20 %) had incomplete but satisfactory hair regrowth.  The treatment was well-tolerated.  The authors concluded that the initial treatment by pulse intravenous corticosteroids may influence the overall response.  They stated that this approach should be evaluated in a larger series of patients.

Acikgoz et al (2014) examined the effect of pulse methylprednisolone therapy for the treatment of adult AA.  Demographic features of all patients were recorded before the treatment.  Patients received methylprednisolone 500 mg intravenously in 3 consecutive days by monthly for 3 months.  Patients were followed-up for 3 months.  Treatment responses were defined by complete regrowth (100 %), significant regrowth (more than 50 %) and minimal regrowth (less than 50 %).  A total of 15 patients enrolled in this study.  At the end of the study, 2 patients had significant regrowth and 1 patient had minimal regrowth in multi-focal AA (n = 4); 1 patient had significant regrowth and 1 patient had minimal regrowth in alopecia universalis (n = 8); 3 patients had no regrowth in alopecia totalis (n = 3).  The authors concluded that these findings suggested that pulse methylprednisolone therapy might be a therapeutic option for severe multi-focal AA.  However, for patients with alopecia totalis or universalis, treatment results were unsatisfactory.  These preliminary findings need to be validated by well-designed studies.

Waldmann (2013) noted that interleukin-15 (IL-15) has a pivotal role in life and death of natural killer (NK) and CD8 memory T cells.  IL-15 signals through a heterotrimeric receptor involving the common gamma chain (γc) shared with IL-2, IL-4, IL-7, IL-9, and IL-21, IL-2/IL-15 receptor β (IL-15Rβ) shared with IL-2 and a private IL-15Rα subunit.  Interferon (IFN)- or CD40 ligand-stimulated dendritic cells coordinately express IL-15 and IL-15Rα.  Cell surface IL-15Rα presents IL-15 in trans to cells that express IL-2/IL-15Rβ and γc. IL-15 is being used to treat patients with metastatic malignancy.  However, IL-15 is an inflammatory cytokine involved in immunological memory including that to self, thereby playing a role in autoimmune diseases.  These insights provided the scientific basis for clinical strategies directed toward diminishing IL-15 action.  Dysregulated IL-15 expression was demonstrated in patients with rheumatoid arthritis, inflammatory bowel disease, psoriasis, celiac disease, and AA.  The monoclonal antibody Hu-Mik-β-1 targets the cytokine receptor subunit IL-2/IL-15Rβ (CD122), blocks IL-15 trans-presentation, and is being used in clinical trials in patients with autoimmune diseases.  In parallel, clinical trials have been initiated involving the Janus kinase-1/2 (Jak1/2) inhibitor ruxolitinib and Jak2/3 inhibitor tofacitinib to block IL-15 signaling.

Hordinsky and Donati (2014) reviewed all randomized controlled trials (RCTs) on the treatment of AA.  These investigators performed a search in the biomedical literature database PubMed, and used the terms “alopecia areata treatment” and article type “randomized controlled trials”.  Following this algorithm, they reviewed, analyzed, and reported on 29 trials that examined the efficacy of anthralin, anti-depressants, biologics, calcineurin inhibitors, corticosteroids (topical and systemic), minoxidil, prostaglandin analogs, sensitizers, and a miscellaneous group of topical and oral drugs with less scientific evidence (aromatherapy, photodynamic therapy, azelaic acid, garlic gel, bexarotene, triiodothyronine, inosiplex, and total glucosides of peony).  The authors concluded that using the American College of Physicians Guideline grading system, their assessment was that the majority of published RCTs of AA were only of moderate quality.  A number of treatments were found to be effective (e.g., topical and oral corticosteroids and the sensitizing agents diphenylcyclopropenone and dinitrochlorobenzene); however, most studies had major limitations that hinder the interpretation of these results.

In a pilot study, Zaher et al (2015) compared the safety and effectiveness of bimatoprost to those of corticosteroid in the treatment of scalp AA.  A total of 30 adult patients with patchy AA (S1) were included.  Two AA patches were randomly assigned to treatment either by mometasone furoate 0.1 % cream once-daily (area A) or bimatoprost 0.03 % solution twice-daily (area B) for 3 months.  Patients were assessed using the SALT scoring system for hair re-growth.  All responding AA patches showed significant reduction in their SALT score after therapy.  Area B demonstrated significantly better results regarding rapidity of response in weeks, percentage of hair re-growth and side effects compared to area A.  The authors concluded that bimatoprost solution represents a therapeutic option for scalp AA. These preliminary findings from a pilot study need to be validated by well-designed studies.

Lux-Battistelli (2015) noted that spontaneous remission occurs in less than 10 % of patients suffering from AA totalis for more than 2 years.  The effectiveness of PUVA therapy is controversial due to recurrence of hair loss after cessation.  These investigators reported 2 cases presenting with AA totalis and AA universalis.  After hair regrowth, relapse of hair loss occurred upon cessation of PUVA and zinc gluconate combination therapy.  However, hair regrowth was noted upon the re-introduction of zinc gluconate and sulfur amino acids without PUVA in the first case and with episodic PUVA in the second case.  The chronology of events appeared to support the notion that zinc has a significant effect.  These findings suggested the possibility of a subgroup of zinc-responsive patients, but the identification of these patients remains difficult.  Metallothioneins and zinc transporters regulating the entrance and exit of zinc in cells might play a key role.  Combination therapy with immunomodulators may be administered to facilitate enhanced zinc-targeted action.  Taking into account the safety profile of zinc, 30 to 40 mg/day of zinc metal may be used during at least 1 year, although these researchers recommend monitoring its serum and hair levels.  The authors concluded that studies with a larger number of patients are needed to further investigate the therapeutic effect of zinc.

An UpToDate review on “Management of alopecia areata” (Messenger, 2015) states that “Further study is necessary to determine whether low-dose recombinant IL-2 therapy should have a role in the treatment of alopecia areata …. Improvement in alopecia totalis during treatment with hydroxychloroquine has been documented in two women with refractory alopecia totalis who were treated with a dose of 200 mg twice daily.  Additional studies are necessary to confirm the efficacy of this therapy.  In our experiences with small numbers of patients, hydroxychloroquine has not been an effective therapy …. A prospective study of 29 adults with alopecia areata involving 40 to 70 % of the scalp suggests that simvastatin/ezetimibe (40 mg/10 mg) may be beneficial for alopecia areata …. A controlled trial is necessary to confirm efficacy of this therapy …. Complete hair regrowth following multiple treatments with a fractional photothermolysis laser has been reported in a patient with alopecia areata refractory to minoxidil and topical and intralesional corticosteroids.  However, further studies are necessary before this approach can be routinely recommended …. Evidence for involvement of neuropeptides in the pathogenesis of alopecia areata and a case report in which alopecia areata associated with neuralgiform head pain improved after botulinum toxin A injection suggested that botulinum toxin might be useful for alopecia areata.  However, additional data to support a beneficial effect are lacking …. Further studies are necessary to determine whether botulinum toxin may be effective for some patients with alopecia areata”.

Acupuncture

Lee and associates (2015) stated that there is no critically appraised evidence of the potential benefits and harm of acupuncture for alopecia areata (AA). This review aims to systematically evaluate the effectiveness of acupuncture for the management of AA in RCTs.  A total of 13 databases will be searched from their inception.  These include PubMed, AMED, EMBASE, the Cochrane Library, 6 Korean medical databases (Korean studies Information Service System, DBPIA, the Town Society of Science Technology, Research Information Sharing Service, KoreaMed and the Korean National Assembly Library), 3 Chinese databases (China National Knowledge Infrastructure Database (CNKI), the Chongqing VIP Chinese Science and Technology Periodical Database (VIP) and the Wanfang Database).  Only RCTs using any type of acupuncture for AA will be considered.  The selection of the studies, data abstraction and validation will be performed independently by 2 researchers.  Methodological quality will be assessed with Cochrane risk of bias.  The systematic review will be published in a peer-reviewed journal.  The review will also be disseminated electronically and in print.  Updates of the review will be conducted to inform and guide the healthcare practice and policy.

Apremilast

Estebanez and colleagues (2019) stated that AA is a common disease characterized by non-scarring hair loss.  There are no satisfactory therapies for extensive cases.  Systemic immune suppressants are usually used despite their non-specific actions and often associated side effects.  Apremilast is an oral, small-molecule, inhibitor of phosphodiesterase 4 approved for the treatment of psoriasis and psoriatic arthritis.  Its use in AA has shown variable results.  Whereas a recent reduced clinical trial concluded a lack of efficacity, several case reports demonstrated a significant improvement.  These investigators reported 4 cases of extensive AA successfully treated with apremilast.  Moreover, they stated that future larger studies are needed to elucidate the role of apremilast in moderate-to-severe AA and examine why some patients respond while others do not.

Brevilin A (Systemic and Topical) for the Treatment of Alopecia Areata

Brevilin A is a sesquiterpene lactone component of Centipeda minima – a traditional Chinese medicinal herb used to treat asthma and cough. In a case-series study, Muscianese and colleagues (2021) examined the safety and efficacy of both systemic and topical brevilin A in refractory AA patients.  After obtaining informed consent, these researchers administered off-label brevilin A to 13 adult patients affected by AA, for a period ranging from 6 to 18 months.  Medical records for each patient and the SALT score before and after brevilin A administration were recorded.  The mean SALT score of these patients was 81.03 (SD 34.9) at baseline and 75.8 (SD 37.4) after brevilin A therapy, meaning no statistically significant improvement was observed (p = 0.2385; paired t-test); however, 3 multi-focal AA (MAA) patients out of 4 achieved an improvement (75 %) suggesting that brevilin A may represent an alternative therapy in this form of AA.  The authors concluded that brevilin A may, in the future, represent a possible effective treatment in patients with multi-focal AA; however, further studies are needed.

Carboxytherapy

Doghaim and colleagues (2018) evaluated the safety and efficacy of carboxytherapy (transcutaneous infusion of carbon dioxide into the affected site) in AA and AGA.  This study was conducted on 80 patients with alopecia divided into 2 groups; Group I included 40 AA patients (Group IA received carboxytherapy and Group IB control received placebo), and Group II included 40 AGA patients (Group IIA received carboxytherapy and Group IIB control received placebo), and followed-up monthly for 3 months.  They were evaluated clinically (by assessment of SALT score in group I, and Sinclair scale and Norwood-Hamilton scale in group II), by dermoscopy and digital dermoscopy at each visit.  Group IA patients showed significant clinical improvement in SALT score and dermoscopic improvement after carboxytherapy and at the end of follow-up period with significant reduction in dystrophic hair, black dots, yellow dots, and tapered hair coinciding with significant emergence of re-growing hair.  Group IIA patients showed significant clinical and dermoscopic improvement after carboxytherapy with significant increase in hair density measured by digital dermoscopy.  However, regression of these results was observed during the follow-up period but was still significantly better than before treatment.  There were statistically significant improvements in clinical score, global assessments, dermoscopic, and digital dermoscopic findings in both group IA and group IIA received carboxytherapy in comparison with group IB and group IIB received placebo injections, respectively.  The authors concluded that carboxytherapy appeared to be a promising therapeutic option for patchy AA and could be helpful as an adjuvant therapy of AGA; but more than 6 sessions are needed and adjuvants are recommended for maintenance of the results.

Chemokine (C-X-C motif) Ligand 1 (CXCL1) and Chemokine (C-X-C motif) Ligand 2 (CXCL2) Gene Polymorphisms Testing

Kim et al (2015a) examined the possible correlations between single nucleotide polymorphisms (SNPs) in the promoter regions of the chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating activity, alpha) (CXCL1) and chemokine (C-X-C motif) ligand 2 (CXCL2) genes and the development of AA in the Korean population. A total of 235 AA patients and 240 control subjects were recruited.  The specific SNPs occurring in the promoter regions of the CXCL1 and CXCL2 genes (rs3117604, -429C/T and rs3806792, -264T/C, respectively) were genotyped.  All data obtained was evaluated using the SNPStats, SPSS 18.0, and the Haploview v.4.2 software platforms.  The odd's ratios (OR), 95 % CI, and p values were calculated using multiple logistic regression models.  Analyses of the genetic sequences obtained revealed a significant correlation between the 2 SNPs and the development of AA (rs3117604, p = 0.0009 in co-dominant model 1, p = 0.01 in co-dominant model 2, p = 0.004 in the dominant model, p = 0.005 in the log-additive model, p = 0.012 in allele distribution; rs3806792, p = 0.036 in co-dominant model 2, p = 0.0046 in the log-additive model).  The TT and CC haplotypes were also observed to show a significant association with increased risk of AA (TT haplotype, p = 0.0018; CC haplotype, p = 0.0349).  The authors concluded that the findings of this study suggested that the CXCL1 and CXCL2 genes may be associated with AA susceptibility.  These preliminary findings need to be validated by further research.

Complementary and Alternative Medicine (CAM) for the Treatment of Alopecia Areata

Tkachenko and colleagues (2023) noted that despite high utilization of complementary and alternative medicine (CAM) for the treatment of AA, the safety and effectiveness of this approach remain unclear.  In a systematic review, these investigators identified all CAM therapies studied for treatment of AA; outcome measures included disease course and psychological well-being.  PubMed and Embase were searched to identify English articles containing original data investigating CAM in human subjects with AA from 1950 to 2018.  Quality was assessed with Oxford Centre for Evidence Based Medicine criteria.  Of 1,015 initial citations, 16 articles met inclusion criteria: 5 RCTs, 5 prospective controlled cohorts, 4 prospective non-controlled cohorts, 1 retrospective cohort, and 1 case series.  CAM therapies with best evidence and efficacy for hair growth in AA included essential oil aromatherapy, topical garlic, and oral glucosides of peony with compound glycyrrhizin.  Hypnosis and mindfulness psychotherapy represented low quality evidence for improvement of psychological and quality of life (QOL) outcomes; adverse events (AEs) were rare and mild for all therapies evaluated.  The authors concluded that this work served to inform physicians that while management of patients with AA seeking CAM is encouraging, further investigation into these therapies to address some of the therapeutic challenges of AA is needed.  Moreover, these researchers stated that inconsistent or poorly reported study methodology and non-standardized outcomes limited the conclusions that could be made from these studies.

Diphencyprone

Lamb et al (2016) noted that contact immunotherapy with diphencyprone (DCP) is used to treat AA; however, its reported effectiveness is variable, and individual response cannot be predicted. These investigators identified patient and treatment course variables that may affect treatment outcome, and reviewed DCP service to identify potential areas for development and improvement.  This study was a retrospective review of a DCP service over a 20-year period (1991 to 2010).  Complete data was available for 205 treatment courses, and 162 (79 %) treatment courses were completed for 133 patient.  Overall, 72.2 % (96/133) of patients had some hair regrowth (any grade).  In 15.8 % of cases (21/133), response was greater than 90 % regrowth. However, 27.1 % (36/133) had no response.  These researchers found that extent of alopecia at baseline and duration of disease were statistically significant when comparing patients with an optimal outcome to those without (p < 0.05).  In contrast to other reports, atopy, age at onset and nail dystrophy were not statistically significant.  For patients receiving more than 1 course, response to DCP treatment was broadly consistent.  The authors concluded that extent of alopecia at baseline and duration of disease were important factors in predicting response.  They stated that the results suggested that atopy should not be considered a predictor of poor outcome with respect to DCP treatment, and there is a need for improved data collection, particularly regarding longer-term outcomes.  Moreover, they stated that the role of maintenance therapy requires objective assessment, and opportunities for DCP self-administration by patients should be explored.  The main drawbacks of this study were its retrospective nature and the lack of long-term follow-up data.

Evaluation of Serum Trace Elements Level

Jin and associates (2017) noted that abnormalities of serum trace elements are involved in the etiology and pathogenesis of alopecia areata; however, the results of published studies are controversial.  In a meta-analysis, these researchers examined the alterations of serum level of trace elements and alopecia areata.  They searched all articles indexed in PubMed, Embase and Science Citation Index published up to April 30, 2016 concerning the association between serum level of zinc, copper, iron/ferritin, selenium or magnesium and alopecia areata.  A total of 10 eligible articles involving 764 subjects were identified.  Overall, pooled analysis indicated that patients with alopecia areata had a lower serum level of zinc (p < 0.001) and selenium (p < 0.001) than the healthy controls.  However, there was no significant difference between the alopecia areata patients and controls in the levels of serum copper (p = 0.81), serum iron (p = 0.36), serum ferritin (p = 0.37) and serum magnesium (p = 0.07).  The authors concluded that the findings of this meta-analysis suggested that low serum levels of zinc and selenium appeared to be important risk factors for alopecia areata.

Furthermore, an UpToDate review on “Clinical manifestations and diagnosis of alopecia areata” (Messenger, 2017b) does not mention measurement of trace elements/selenium/zinc as a diagnostic tool. Guidelines from the British Association of Dermatology on alopecia areata (Messenger, et al., 2012) state that Investigations are unnecessary in most cases of alopecia areata.: "One small case series suggested that iron deficiency is more common in women with alopecia areata than the population at large but this was not confirmed in two subsequent studies, and routine testing for iron status is not recommended. There are no published studies demonstrating a treatment response to iron replacement therapy."

Excimer Laser

Gundogan et al (2004) described the use of the excimer laser in 2 patients with alopecia areata with evidence of hair regrowth and good tolerability.  However, these investigators stated that this new means of treatment has yet to be discussed in medical literature.  The investigators concluded that large prospective studies are needed to evaluate the potential clinical value of the excimer laser in treating alopecia areata.

Al-Mutairi (2007) stated that AA is loss of hair from localized or diffuse areas of hair-bearing area of the skin.  Recently, there were reports of effectiveness of the 308-nm excimer radiation for this condition.  These researchers examined the effect of the 308-nm excimer laser in the treatment of AA.  A total of 18 patients with 42 recalcitrant patches (including 1 adult with alopecia totalis) were included in this study.  The lesions were treated with the 308-nm excimer laser twice-weekly for a period of 12 weeks; 1 lesion on each patient was left as a control for comparison.  There were 7 males and 11 females in this study.  Re-growth of hair was observed in 17 (41.5 %) patches; 13 of the 18 lesions in scalp showed a complete re-growth of hair.  The extremity regions failed to show a response.  Atopic diatheses had an unfavorable effect on the outcome in this cohort of patients.  The authors concluded that the 308-nm excimer laser was an effective therapeutic option for patchy AA of the scalp and for some cases with patchy AA of the beard area.  It did not work for patchy AA of the extremities.  This was a small study (n = 18 [1 with alopecia totalis])

Mehraban and Feily (2014) stated that 308nm xenon-chloride excimer laser, a novel mode of phototherapy, is an ultraviolet B radiation system consisting of a noble gas and halide. The aim of this systematic review was to investigate the literature and summarize all the experiments, clinical trials and case reports on 308-nm excimer laser in dermatological disorders.  308-nm excimer laser has currently a verified efficacy in treating skin conditions such as vitiligo, psoriasis, atopic dermatitis, alopecia areata, allergic rhinitis, folliculitis, granuloma annulare, lichen planus, mycosis fungoides, palmoplantar pustulosis, pityriasis alba, CD30+ lympho proliferative disorder, leukoderma, prurigo nodularis, localized scleroderma and genital lichen sclerosus.  The authors concluded that although the 308-nm excimer laser appears to act as a promising treatment modality in dermatology, further large-scale studies should be undertaken in order to fully affirm its safety profile considering the potential risk, however minimal, of malignancy, it may impose.

Byun et al (2015) noted that some studies have reported the use of 308-nm excimer laser therapy for the treatment of AA; however, the effectiveness of this therapy on a theoretical basis has not yet been comparatively analyzed.  These researchers examined the therapeutic effect of excimer laser therapy on AA.  One alopecic patch was divided into control and treated sides in 10 patients with AA.  Then, 308-nm excimer laser therapy was administered twice-weekly for 12 weeks.  Photograph and phototrichogram analyses were carried out.  Photographic assessments by both dermatologists and individuals of the general population showed objective improvements after excimer laser therapy.  On the treated side, the hair count and hair diameter had statistically increased after treatment.  However, only the hair diameter was found to be significantly high in the treated half when it was compared with the control side.  The authors concluded that the 308-nm excimer laser had a therapeutic effect on AA, which was proven by photograph and phototrichogram analysis by a side-by-side comparison.  This was a small study (n = 10).

Lee et al (2020) stated that excimer laser/light (EL) has been reported to be effective for the treatment of AA, but its response has not been systematically reviewed.  In a systematic review and meta-analyses, these researchers examined the safety and treatment response of EL treatment of AA.  They carried out a comprehensive search of the Medline, Embase, Cochrane library, and Web of Science (from inception to December 31, 2018) to identify prospective clinical studies examining the treatment response of EL for AA.  The primary outcome was cosmetically acceptable hair re-growth (hair re-growth greater than or equal to 75 %); random-effects meta-analyses using generic inverse variance weighting were performed to estimate treatment responses.  Of 52 records initially identified, 13 full-text articles were finally evaluated in terms of eligibility.  A total of 9 prospective clinical studies (129 AA patients) including 5 controlled clinical trials were identified.  Cosmetically acceptable hair re-growth was achieved in 50.2 % (95 % CI: 31.5 % to 68.9 %; 8 studies).  EL treatment significantly improved hair re-growth compared with untreated controls (RR 7.83; 95 % CI: 2.11 to 29.11; 5 controlled clinical trials).  No serious adverse effect was noted.  The authors concluded that EL treatment appeared to produce a favorable therapeutic response in AA patients.  These researchers stated that the use of EL should be encouraged for AA patients with the advantages of the non-invasiveness and no systemic effect.

Gupta and Carviel (2021) noted that AA is an autoimmune disease that can result in spontaneous hair loss.  Currently, there is no Food and Drug Administration (FDA)-approved treatment, however new treatments are being examined.  Excimer laser and excimer lamp treatment have been suggested and have the benefit of mild/few adverse effects.  These researchers carried out a literature search and meta-analysis to examine the effectiveness of the Excimer laser and lamp for treatment of AA.  No controlled trials were found that use the 308-nm Excimer lamp; 4 controlled trials (n = 105) testing the effectiveness of the 308-nm excimer laser were identified.  When laser treatment was compared to control measured via the number of responders to treatment, the standardized mean difference (SMD) was 18.37 (95 % CI: 3.28 to 102.77) in favor of treatment (p < 0.0009, I2 = 36 %).  The authors concluded that these findings suggested that use of the 308-nm Excimer laser can be effective in AA therapy; however, more studies are needed observing both the 308-nm Excimer laser and lamp.

Furthermore, an UpToDate review on “Alopecia areata: Management” (Messenger, 2022) states that “Improvement following treatment with a variety of additional local and systemic therapies has been reported.  However, further studies are necessary before any of these agents can be routinely recommended … The excimer laser emits monochromatic ultraviolet B (UVB) light at a wavelength of 308 nm.  Its mechanism of action in alopecia areata is thought to involve the induction of T cell apoptosis.  In a few small studies and case reports, treatment with the excimer laser was associated with improvement in patchy alopecia areata of the scalp.  Patients with lesions on the extremities, alopecia totalis, or alopecia universalis have not responded to therapy”.

FAS/FASL, IL2RA and CTLA4 Genetic Polymorphisms Testing for Alopecia Areata

Gil-Quinones and colleagues (2021) noted that genetic association studies on AA carried out in various populations have shown heterogeneous results.  In a systematic review and meta-analysis, these investigators examined the results of said studies to estimate the impact of FAS, FASL, PTPN22, CTLA4 and IL2RA gene polymorphisms on AA susceptibility.  They carried out a systematic literature search in the Medline, Web of Science, Scopus, Embase and LILACS databases.  Studies published up to June 2020 were included.  The results available in the grey literature including the Open Grey and Google Scholar databases were also used.  The texts of potentially related studies were screened by individual reviewers.  Evidence of publication bias was assessed using the Newcastle-Ottawa scale and the quality of evidence was assessed using the Grading of Recommendations, Assessment, Development and Evaluation (GRADE) system.  The quantitative synthesis was performed using the fixed effect model.  Out of 1,784 articles, these researchers identified 18 relevant articles for the qualitative synthesis and 16 for the quantitative synthesis.  In a study of rs2476601 polymorphism of PTPN22 gene, including 1,292 cases and 1,832 controls, a correlation was found with the risk of developing AA in the allelic model (OR 1.49; 95 % CI: 1.13 to 1.95]), the heterozygous codominant (OR 1.44; 95 % CI: 1:19 to 1.76]) and dominant model (OR 1.43; 95 % CI: 1.18 to 1.73).  No association was found between the presence of FASL, PTPN22, CTLA and IL2RA gene polymorphisms with AA susceptibility.  The authors concluded that the findings of this systematic review and meta-analysis suggested that the T allele of the single nucleoid polymorphism (SNP) rs2476601 in PTPN22 gene was a risk factor for developing AA.  However, these researchers stated that more robust studies defining the ethnic background of the population of origin are needed, so that the risk identified in the present study can be validated.  In addition, further investigation is needed to examine the role of the FAS, FASL, PTPN22, CTLA4 and IL2RA genetic variants, given the heterogenous results found in the literature.

The authors stated that the outcomes obtained in this study revealed several drawbacks that must be considered for data interpretation.  Regarding the PTPN22 gene, it was not possible to analyze the subgroups by ethnicity, which usually allows a better interpretation of the data of multi-factorial diseases such as AA.  In addition, it was only possible to carry out a meta-analysis for 1 variant per gene from the data obtained in the extraction phase, since not all studies analyzed the association between AA and the same gene polymorphisms.  For that reason, these researchers could not identify the contribution of each variant to the susceptibility to develop the disease.  Haplotype analysis may have provided more information regarding the role played by the variants in AA risk.  On the other hand, the number of studies included for the combined analysis of each gene was limited, which reduced the statistical power of the estimates and allowed these investigators to identify that the estimates could be altered due to publication bias.  Some of the analyses proposed could not be performed since all the included studies did not consider the same variables, such as the different forms of AA.

Fractional Carbon Dioxide Laser

In a case-series study, Majid and colleagues (2018) examined the safety and efficacy of a combination of fractional carbon dioxide (CO2) treatment followed by topical corticosteroid application in resistant AA.  A total of 10 cases of resistant AA who had not responded to multiple treatment modalities were treated with fractional CO2 laser followed by topical application of triamcinolone spray (10 mg/ml) on the resistant lesions.  Patients received 4 to 8 sessions that were repeated at an interval of 3 to 4 weeks.  Response to treatment was assessed on a quartile physician assessment scale and labeled as excellent (greater than 75 % regrowth), good (50 % to 75 % re-growth), fair (26 % to 50 % response), and poor (less than 25 % re-growth); 8 of these 10 cases completed the treatment process; 7 of the 8 patients had complete recovery of the area treated; 1 patient however did not show good response even after 4 sessions.  No significant adverse effects were noted in any of the patients.  The authors concluded that fractional CO2 laser in combination with topical triamcinolone could prove to be an effective therapeutic option in resistant AA.  These researchers stated that the limitations of this study were: This was a case-series study conducted in a limited number of patients (n = 10), and larger studies with more patients are needed to confirm these preliminary findings.

Nouch (2022) noted that fractional CO2 laser and cryotherapy are used to stimulate hair growth in patients of AA.  These investigators examined the effectiveness of fractional CO2 laser versus liquid nitrogen cryotherapy in the treatment of AA; participants were 80 patients presented with patches of AA.  The 1st group consisted of 40 patients who were treated with fractional CO2 laser, and the 2nd group was also 40 patients who were treated with cryotherapy.  The effectiveness of the treatment was statistically evaluated.  A positive therapeutic response was noted in most patients by trichoscopy at the end of treatment, there was a decrease in exclamation mark hairs, yellow dots, black dots, broken hairs, and an increase in short vellus hair and circle hair of patients compared to baseline and control.  No serious side effects were observed.  The authors concluded that fractional CO2 laser and liquid nitrogen cryotherapy are considered a promising, safe, and effective treatment modality with the better result with CO2 laser.

HLA-DRB1 Polymorphisms and Alopecia Areata Disease Risk

Ji and colleagues (2018) stated that published studies have reported conflicting and heterogeneous results regarding the association between human leukocyte antigen (HLA)-DRB1 polymorphisms and AA.  These researchers quantitatively analyzed the association between HLA-DRB1 polymorphisms and AA.  In this study, all relevant publications were searched through December 2016; ORs and CIs for comparisons between case and control groups were calculated.  Stata 14.0 software was used to perform statistical analysis.  A total of 12 articles were identified.  For HLA-DRB1*04 and HLA-DRB1*16 polymorphisms, the OR (95 % CIs) was 1.49 (1.24 to 1.78) and 1.61 (1.08 to 2.41), and p was < 0.01 and < 0.01, respectively.  For HLA-DRB1*0301, HLA-DRB1*09, and HLA-DRB1*13 polymorphisms, the OR (95 % CIs) was 0.42 (0.28 to 0.63), 0.74 (0.55 to 0.99), and 0.62 (0.40 to 0.98), and p was < 0.01, < 0.01, and < 0.01, respectively.  Statistical evidence revealed no publication bias (p >  0.05).  The authors concluded that the present meta-analysis suggested that HLA-DRB1*04 and HLA-DRB1*16 polymorphisms might be associated with increased AA risk, while HLA-DRB1*0301, HLA-DRB1*09, and HLA-DRB1*13 polymorphisms might decrease the AA risk.  These investigators stated that studies with adequate methodological quality on gene-gene and gene-environment interactions are needed to validate the results in the future.

The authors stated that to avoid local literature bias, they obtained and included both English and Chinese language reports.  And yet, some shortcomings of the analysis could not be neglected.  First, the number of included studies was limited because the incidence of HLA-DRB1 genotypes was low.  Enough information could not be obtained on clinical type and magnitude for subgroup analysis due to the limited number of included studies.  Second, it was uncertain whether the cases were comparably representative, although significant publication bias between studies was not detected.

Inflammatory Biomarkers for Alopecia Areata

Islamoglu and Demirbas (2020) stated that AA is a chronic inflammatory disease characterized by non-cicatricial hair loss; and the cause of the disease is still unknown.  It could appear at any age and occurs in 0.2 % of the general population.  Red cell distribution width (RDW), mean platelet volume (MPV), plateletcrit (PCT), the ratio of neutrophils to lymphocytes (NLR), and the ratio of platelets to lymphocytes (PLR) have all been reported as inflammatory markers in recent studies.  However, these parameters have not been examined in AA patients.  In a retrospective study, these investigators examined the hematological and inflammatory parameters of AA and considered their association with disease severity.  A total of 105 patients with AA and 108 healthy controls were enrolled in the study; RDW, MPV, PCT, NLR, PLR, erythrocyte sedimentation rate (ESR), and C-reactive protein (CRP) were evaluated.  Disease severity was assessed by using the Severity of Alopecia Tool (SALT) score.  There were no statistically significant differences in RDW, MPV, PCT, NLR, and PLR levels between AA patients and healthy controls; only CRP values were significantly higher in the AA group.  The authors concluded that complete blood count (CBC) parameters are low-cost tests that can be used to define inflammation levels in inflammatory diseases.

Dere and Gundogdu (2021) noted that AA is an inflammatory condition that causes regional shedding without scarring on the hair or eyebrows, eyelashes, and beard.  Previous studies had reported that MPV, NLR, and PLR are significant indicators of systemic inflammation in various diseases.  In a retrospective study, these researchers examined these parameters in AA patients and assessed their relationship with the disease.  A total of 135 AA patients and 135 healthy controls who did not have any systemic, inflammatory, infectious, or autoimmune diseases were included in this study.  Demographic characteristics and CBC results were analyzed, and statistical analysis was carried out; PLR, NLR, and MPV values were not different between AA patients and the control group.  The authors concluded that PLR, NLR, and MPV values are not convenient parameters to show inflammatory response in AA.

Furthermore, an UpToDate review on “Alopecia areata: Management” (Messenger, 2021) does not mention the use of inflammatory biomarkers as a management tool. 

Interleukin-12, Interleukin-17, Interleukin-18, and Interleukin-23 Receptor Gene polymorphisms Testing

In a case-control study, Aytekin et al (2015) examined the distribution of interleukin (IL)-12 (IL12; 1188A/C), IL17 (A7488G) and IL-23 receptor (IL23R; +2199A/C) gene polymorphisms in patients with AA. Patients with AA and healthy controls were enrolled in this study.  Genotyping of the IL12 (1188A/C), IL17 (A7488G) and IL23R (+2199A/C) polymorphisms was carried out.  Genotype frequencies were compared between the 2 groups.  The study enrolled 100 patients with AA and 71 control subjects.  No significant differences were found in the frequencies for the IL12 and IL23R gene polymorphisms between the patient and control groups.  The IL17 GG genotype was significantly more common and the IL17 GA genotype was significantly less common in patients with AA compared with controls, but only 10 % of patients had the GG genotype.  The authors concluded that IL17 GG genotype was associated with susceptibility for AA, but this genotype was only present in a small number of patients.  Moreover, the IL12 and IL23R gene polymorphisms were not found to have a significant association with AA.

Celik and Ates (2018) examined if the IL-18 (rs187238 and rs1946518) SNPs may be associated with AA and/or clinical outcome of patients with AA in Turkish population.  Genotyping of rs187238 and rs1946518 SNPs were detected using sequence-specific primer-polymerase chain reaction (SSP-PCR) method in 200 patients with AA and 200 control subjects.  The genotype distribution of rs1946518 (-607C>A) SNP was found to be statistically significantly different among patients with AA and controls (p = 0.0008).  Distribution of CC+CA genotypes and frequency of -607/allele C of rs1946518 SNP were higher in patients with AA (p = 0.001, p = 0.001, respectively).  The genotype distribution of rs187238 (-137G>C) SNP was found to be statistically significantly different among patients with AA and control subjects (p = 0.0014).  Distribution of GG genotype and frequency of -137/allele G of rs187238 SNP were higher in patients with AA (p = 0.0003, p = 0.001, respectively).  The authors concluded that rs1946518 (-607C>A) and rs187238 (-137G>C) polymorphisms were found to be associated with AA.  They stated that the findings of this study suggested that IL-18 rs187238 and rs1946518 SNPs may be the cause of the AA susceptibility.

Intralesional Candida Antigen Immunotherapy

Ali and colleagues (2021) noted that AA is an autoimmune hair follicle disorder that is challenging to treat.  Although there are multiple topical immunotherapeutic agents, their side effects limit their use.  Candida antigen can serve as another immunotherapeutic treatment, with an easier application and fewer side effects.  These researchers examined the efficacy of Candida antigen versus intralesional steroids for the treatment of AA.  A total of 48 adult patients with AA were divided into 2 groups: the Candida group included 24 patients who were injected monthly with 0.1-ml of Candida albicans antigen in 1 patch of alopecia, and the intralesional corticosteroid group included 24 patients who were injected monthly with 0.1-ml of triamcinolone acetonide, as a control group, in all alopecia patches.  After 5 months, there was a highly significant decrease in the severity of AA in both groups with no significant difference between them.  In the Candida group, 8 patients showed complete hair regrowth and 9 patients showed partial regrowth.  Side effects were mild and transient such as pain during injection, which was significantly lower in the Candida group than in the corticosteroid group.  The authors concluded that intralesional Candida antigen is a promising treatment for AA with differing severities.  These preliminary findings need to be validated by well-designed studies.

Furthermore, an UpToDate review on “Alopecia areata: Management” (Messenger, 2021) does not mention Candida antigen immunotherapy as a management / therapeutic option. 

Intralesional Methotrexate

In a randomized, clinical trial, Hamdino and colleagues (2022) compared the safety and effectiveness of intralesional methotrexate (MTX) versus triamcinolone acetonide (TrA) in the treatment of localized AA in adults, both clinically and trichoscopically.  A total of 40 adult patients with localized AA were recruited and divided into 2 groups: 20 patients were treated by intralesional TrA and the other 20 patients were treated by intralesional MTX every 3 weeks, for maximum 4 sessions.  These researchers carried out clinical and trichoscopic examinations at baseline, each session and for 3 months after the last session.  At the end of sessions (12 weeks), regrowth scale was significantly higher in TrA group compared to MTX group (p = 0.028); however, at 3-month follow-up, regrowth scale was higher in MTX group compared to TrA group (p = 0.153).  There was a statistically significant reduction in AA-specific trichoscopic signs after 12 weeks and at the 3-month follow-up in both groups.  Local AEs in both groups were transient and disappeared during the follow-up period.  The authors concluded that intralesional MTX in treatment of localized AA in adults could be promising and comparable to intralesional TrA with the need for further controlled and extensive trials.  Trichoscopy could reveal early clinical response via disappearance of AA-specific trichoscopic signs and also early detection of adverse effects.

Low-Level Laser Therapy

Zarei and colleagues (2016) noted that despite the current treatment options for different types of alopecia, there is a need for more effective management options. Recently, low-level laser therapy (LLLT) was evaluated for stimulating hair growth.  These investigators reviewed the current evidence on the LLLT effects with an evidence-based approach, focusing more on RCTs by critically evaluating them.  In order to examine if in individuals presenting with hair loss (male pattern hair loss (MPHL), female pattern hair loss (FPHL), AA, and chemotherapy-induced alopecia (CIA)) LLLT is effective for hair regrowth, several databases including PubMed, Google Scholar, Medline, Embase, and Cochrane Database were searched using the following keywords: alopecia, hair loss, hair growth, low level laser therapy, low level light therapy, low energy laser irradiation, and photobiomodulation.  From the searches, a total of 21 relevant studies were summarized in this review including 2 in-vitro, 7 animal, and 12 clinical studies.  Among clinical studies, only 5 were RCTs, which evaluated LLLT effect on male and female pattern hair loss.  The RCTs were critically appraised using the created check-list according to the Critical Appraisal for Therapy Articles Worksheet created by the Center of Evidence-Based Medicine, Oxford.  The results demonstrated that all the performed RCTs had moderate-to-high quality of evidence.  However, only 1 out of 5 studies performed intention-to-treat analysis, and only another study reported the method of randomization and subsequent concealment of allocation clearly; all other studies did not include this very important information in their reports.  None of these studies reported the treatment effect of factors such as number needed to treat.  Based on this review on all the available evidence about effect of LLLT in alopecia, the authors found that the FDA-cleared LLLT devices are both safe and effective in patients with MPHL and FPHL who did not respond or were not tolerant to standard treatments.  They stated that future RCTs of LLLT are strongly encouraged to be conducted and reported according to the Consolidated Standards of Reporting Trials (CONSORT) statement to facilitate analysis and comparison.

Manganese Superoxide Dismutase (MnSODAla-9Val) and Glutathione Peroxidase (GPx1 Pro 197 Leu) Gene Polymorphisms Testing

Kalkan et al (2015) stated that the role of the oxidative stress in AA has been studied by several researchers in a few studies with conflicting results. These results suggested that lipid peroxidation and alterations in the oxidant-antioxidant enzymatic system may play a role in the pathogenesis of AA.  These researchers examined the possible associations between the MnSOD Ala-9Val and GPx1 Pro 198 Leu polymorphisms and AA susceptibility and disease progression in Turkish population.  The study group consisted of 119 unrelated patients with AA and 104 unrelated healthy controls with no scalp lesions in their personal history or on clinical examination.  Genotyping was performed to identify MnSOD Ala-9Val and GPx1 Pro 198 Leu polymorphisms by a method based on polymerase chain reaction (PCR) amplification and detection of polymorphisms with hybridization probes labeled with fluorescent dyes.  Genotype and allele frequencies were compared between patients with AA and healthy control subjects.  There was no significant difference between the MnSOD Ala-9Val SNP genotype distributions and allele frequencies of the AA patients and the control group (p = 0.168 and p = 0.820, respectively).  There was not any association between clinical and demographical features of the study patients with AA and MnSOD Ala-9Val and GPx1 Pro 198 Leu polymorphism genotypes except gender.

Measurements of Serum Levels of Interleukins 10, 12, 17 and 35 for Alopecia Areata

Wojciechowska-Zdrojowy and colleagues (2021) examined the concentration of IL-10, IL-12, IL-17 and IL-35 in the blood serum of patients with AA.  The study group (A patients) consisted of 118 patients.  The control group consisted of 54 healthy individuals.  The severity of the disease was assessed by SALT score.  In the study group, the percentage of hair loss averaged 49.6 % ± 36.3 %.  The concentration of IL-10, IL-12, IL-17 and IL-35 in the serum was assessed by the enzyme-linked immunosorbent assays (ELISA).  In patients with AA, the level of IL-12 and IL-17 was significantly higher than in the control group (p > 0.05).  The level of IL-10 in patients was slightly higher, whereas the level of IL-35 was slightly lower, compared to the control group; however, those differences were not statistically significant.  In addition, in patients with more severe disease the IL-12 level was significantly higher as compared to patients with the less severe AA (p < 0.05).  The authors concluded that the etiopathogenesis of AA is complex, however Th1 and Th17 lymphocytes and their increased activity are undoubtedly significant contributors in this process.  Moreover, these researchers stated that disorders of immunological processes in AA require further research to understand the underlying pathomechanisms of the disease and to provide potential therapeutic strategies.

Mesenchymal Stem Cells for the Treatment of Alopecia Areata

Czarnecka and colleagues (2021) stated that AA is the 2nd most common cause of non-scarring alopecia.  Little is known on the etiopathogenesis of AA.  It is considered an autoimmune disease, with T lymphocytes and antibodies directed against hair follicle structures.  Topical and systemic therapies are used for the treatment of AA; however, none of the therapies used to-date exhibit a permanent therapeutic effect.  These researchers examined the safety and efficacy of AA treatment via a single intra-dermal injection of a suspension of allogeneic mesenchymal stem cells (MSCs) extracted from Wharton's jelly (WJ-MSCs) into the alopecia foci.  The study included 4 AA patients who underwent experimental therapy with a suspension of WJ-MSCs.  The AA intensity was measured using the SALT score.  This measure was carried out 3 times during treatment: 1st measure (SALT0) prior to treatment; 2nd measure (SALT12) 12 weeks after the treatment; and 3rd measure (SALT24) 24 weeks after the treatment.  Furthermore, during each follow-up visit (6, 12, 18, and 24 weeks after the administration of WJ-MSCs) the patient's general condition (physical examination) and local condition were evaluated, their mood was examined, and a photo of the scalp was taken.  Hair regrowth was observed in all patients by an average of 67 % at the sites where the cell suspension was administered.  In all cases, these investigators observed greater dynamics of hair regrowth in the first 3 months following the treatment, with an average increase of 52.2 %, compared to the following 3 months, with an average of 32 %.  The authors concluded that the results of the applied intra-dermal injections of an allogeneic WJ-MSC suspension were positive with hair growth observed in all subjects, and the therapy was found to be safe, with no side effects.  The main drawback of this study was the small number of patients enrolled (n = 4).

The authors noted that to their knowledge, this was the 1st clinical study to describe the use of an allogeneic MSC transplant in patients with AA.  The results of this treatment were positive, with hair growth observed in all subjects, and the therapy was found to be safe, with no side effects.  These investigators stated that the question remains as to how many cells should be applied to the patient to achieve full hair regrowth and how often the treatments should be repeated to achieve 100 % therapy efficacy with no relapse.  These researchers emphasized the need to carry out further studies with a randomized control group.

Furthermore, an UpToDate review on “Alopecia areata: Management” (Messenger, 2021) does not mention mesenchymal stem cell as a management / therapeutic option.

Minoxidil

In a systematic review and meta-analysis, Freire and colleagues (2019) examined the safety and effectiveness of over 80 interventions for AA, including minoxidil -- one of the most promising interventions for patchy AA in children and adults of both sexes.  These investigators carried out an extensive search of international medical literature involving RCTs of AA interventions; RCTs were evaluated qualitatively and quantitatively according to the previously published protocol and for 7 specific outcomes.  The meta-analysis involving 5% minoxidil versus placebo presented a significant difference in favor of 5 % minoxidil with the moderate quality of evidence in children and adults with patchy AA (relative risk [RR] 8.37; 95 % CI: 3.16 to 22.14); no severe adverse event (AE) was reported.  The authors concluded that treatment of patchy AA with 5 % minoxidil proved effective, and clinically and statistically safe in studies with limited sample size; quality of evidence was moderate.  These researchers stated that further studies with sound methodological quality, more subject and outcome observations lasting longer than 6 months are needed to address remaining uncertainties.

Platelet-Rich Fibrin for the Treatment of Alopecia Areata

Vazquez and colleagues (2022) noted that PRP has been shown to release a multitude of growth factors; however, its preparation requires the use of anti-coagulants.  In contrast, advanced platelet-rich fibrin (aPRF) is produced by centrifuging whole blood and allowing it to clot.  The clot contains the platelets, growth factors, and neutrophils, and it is composed of a fibrin matrix that continuously releases more growth factors over a longer time period.  Advanced PRF is commonly used in dental and bone grafting procedures, but it is rarely used for cosmetic injectables because its high density makes it difficult to inject through smaller gauge needles.  These researchers described a technique whereby aPRF was reduced to an injectable form with micronization; it was used to treat AA in a 28-year-old man who developed it after symptomatic COVID-19 infection a month before presentation.  The subject was vaccinated in between infection, and symptoms were limited to headache and sore throat.  He had complete resolution of his AA at 6-month follow-up with only 2 treatments as opposed to monthly intralesional steroids.  The authors reported these findings using aPRF for AA with promising results as a possible future treatment for patients with this autoimmune disease.  This was a single-case study; its findings need to be validated by well-designed studies.  In addition, more research is needed to prove AA as a probable result of COVID-19 infection.

Platelet-Rich Plasma

In a randomized, double-blind, placebo- and active-controlled, half-head, parallel-group study, Trink et al (2013) evaluated the safety and effectiveness of platelet-rich plasma (PRP) for the treatment of AA.  A total of 45 patients with AA were randomized to receive intralesional injections of PRP, triamcinolone acetonide (TrA) or placebo on one half of their scalp.  The other half was not treated.  Three treatments were given for each patient, with intervals of 1 month.  The end-points were hair regrowth, hair dystrophy as measured by dermoscopy, burning or itching sensation, and cell proliferation as measured by Ki-67 evaluation.  Patients were followed for 1 year.  Platelet-rich plasma was found to increase hair regrowth significantly and to decrease hair dystrophy and burning or itching sensation compared with TrA or placebo.  Ki-67 levels, which served as markers for cell proliferation, were significantly higher with PRP.  No side-effects were noted during treatment.  The authors concluded that the findings of this pilot study, which was the first to investigate the effects of PRP on AA, suggested that PRP may serve as a safe and effective treatment option in AA, and calls for more extensive controlled studies with this method.

Also, an UpToDate review on “Management of alopecia areata” (Messenger, 2014) states that “Platelet-rich plasma, which contains growth factors that are important for cell proliferation and differentiation and has antiinflammatory properties, may be beneficial in alopecia areata.  In a trial in which 45 patients with chronic recurring alopecia areata of at least two years duration were randomly assigned to intralesional injections of autologous platelet-rich plasma, triamcinolone acetonide, or placebo administered once per month for three months, platelet-rich plasma injection was most effective for inducing hair regrowth.  Platelet-rich plasma therapy also was associated with reductions in symptoms of burning or itching in affected areas.  Additional studies are necessary to validate the findings of this trial”.  Furthermore, the review does not mention the use of IL-15 blockers (e.g., ruxolitinib and tofacitinib) as therapeutic options.

Ayatollahi and colleagues (2017) noted that although there are many studies showing the role of PRP in bone grafts, teeth osteosynthesis, and wound healing, there have been little peer-reviewed studies about the safety and efficacy of PRP application in the treatment of hair loss.  In this systematic review, these investigators searched Ovid Medline, Scopus and Web of Knowledge till November 2015 for human studies evaluating the effectiveness of PRP for the treatment of non-cicatricial alopecia.  Among 704 papers retrieved in first search, 18 papers matched the inclusion criteria, 14 for androgenic alopecia and 4 for alopecia areata.  They included 2 case reports, 8 case series, 6 controlled clinical trials, and only 2 RCTs.  The authors concluded that most of the available evidence has shown low quality and controversial results about the effectiveness of PRP in treating non-cicatricial alopecias, including androgenetic alopecia and alopecia areata.  They stated that further RCTs with larger sample size and standard protocols regarding the number and interval of treatment sessions, number of platelets, method of activation, etc., are needed to examine and safety and effectiveness of PRP in treating hair loss.

Furthermore, an UpToDate review on “Management of alopecia areata” (Messenger, 2017a) states that “Platelet-rich plasma, which contains growth factors that are important for cell proliferation and differentiation and has anti-inflammatory properties, may be beneficial in alopecia areata.  In a trial in which 45 patients with chronic recurring alopecia areata of at least 2 years duration were randomly assigned to intralesional injections of autologous platelet-rich plasma, triamcinolone acetonide, or placebo administered once per month for 3 months, platelet-rich plasma injection was most effective for inducing hair regrowth.  Platelet-rich plasma therapy also was associated with reductions in symptoms of burning or itching in affected areas.  Additional studies are necessary to validate the findings of this trial”.

Cervantes and colleagues (2018) evaluated the effectiveness of PRP treatment for androgenetic alopecia (AGA).  A total of 12 studies conducted from 2011 to 2017 were evaluated and summarized by study characteristics, mode of preparation, and treatment protocols.  A total of 295 subjects were given PRP or control treatment in these studies, and evaluated for terminal hair density, hair quality, anagen/telogen hair ratio, keratinocyte proliferation, blood vessel density, etc.  Some studies also provided subject self-assessment reports.  Most of the studies reviewed showed effectiveness of PRP in increasing terminal hair density/diameter.  The authors concluded that additional investigations are needed to determine the optimal treatment regimen for high efficacy of PRP in AGA.

In a meta-analysis, Giordano and associates (2018) compared local injection of PRP versus control to examine the efficacy of local PRP injections in AGA.  These investigators performed a literature search.  The increase in number of hairs was the primary outcome; secondary outcomes were the increase of hair thickness and the percentage increase in hair number and thickness.  A total of 7 studies involving 194 patients were retrieved and included in the present analysis.  A significantly locally increased hair number per cm2 was observed after PRP injections versus control (mean difference [MD] 14.38, 95 % CI: 6.38 to 22.38, p < 0.001).  Similarly, a significantly increased hair thickness cross-section per 10-4 mm2 (MD 0.22, 95 % CI: 0.07 to 0.38, p = 0.005) favoring PRP group.  The pooled results did not show a significant percentage increase in hair number (MD 18.79 %, 95 % CI: 8.50 to 46.08, p = 0.18), neither hair thickness (MD 32.63 %, 95 % CI: 16.23 to 81.48, p = 0.19) among patients treated with PRP.  The authors concluded that local injection of PRP for AGA might be associated with an increased number of hairs in the treated areas with minimal morbidity, but there is clearly a lack of scientific evidence on this treatment modality.  They stated that further studies are needed to evaluate the efficacy of PRP for AGA.

In a randomized, placebo-controlled, split-head, pilot study, Gupta and colleagues (2021) examined the effect of PRP on hair regrowth and lesional T-cell cytokine expression in AA.  This trial enrolled 27 patients with AA (SALT score of greater than or equal to 25 %).  Alopecia patches on either side of the scalp were randomized to receive 3 intra-dermal injections of PRP or normal saline at monthly intervals and examined 3 months after the last session.  Lesional T-cell cytokine messenger RNA expression was compared pre- and post-treatment in the PRP-treated sites.  The mean SALT score did not change significantly compared with baseline with either PRP or placebo injections at any visit; however, the mean percentage reduction in the score in the PRP arm was more than in the placebo arm (9.05 % ± 36.48 % versus 4.99 % ± 33.88 %; p = 0.049) at final assessment.  The mean IFN-gamma (p = 0.001) and IL-17 cytokine (p = 0.009) messenger RNA expression decreased, whereas the mean IL-10 (p = 0.049) and FOXP3 (p = 0.011) messenger RNA expression increased significantly after PRP treatment.  The authors concluded that PRP was found to have limited efficacy in AA; however, it may play a role in restoring immune balance in the alopecic patches.

In a systematic review and meta-analysis, Cruciani et al (2023) examined the benefit of PRP in the treatment of alopecia.  These investigators searched Medline (through PubMed), Embase, and CENTRAL for relevant data.  Treatment effect was described by mean difference (MD) and risk difference with 95 % CI.  The GRADE system was used to assess the certainty of the body of evidence.  They found 27 controlled trials (1,117 subjects) that met inclusion criteria: 18 trials (713 subjects) in patients with AGA, and 9 (404 subjects) in patients with AA; 11 studies had a split head design.  There was heterogeneity in types of PRP (e.g., activated and non-activated) and administration schedules.  PRP was compared to saline injections (18 studies), local steroid injections (4 studies) and other comparators (5 studies).  Most commonly reported outcomes were hair density and hair re-growth.  It was not possible to pool all outcome data because of heterogeneity in reporting, and because reporting was often limited to a single study.  Compared to saline injections, PRP injections increased hair density over a medium-term follow-up (MD, 25.6 hairs/cm2; 95 % CI: 2.62 to 48.57); however, the evidence was rated as low quality due to inconsistency and risk of bias.  In individuals with AA, it is unclear whether PRP injection compared with triamcinolone injection increase the rate of subjects with hair re-growth (very-low quality of evidence due to inconsistency, imprecision, and risk of bias).  There were no serious AEs related to PRP injection or control treatments.  The authors concluded that there is limited evidence showing benefit of PRP for treatment of alopecia, and most of this evidence was of low quality.

Prostaglandins (e.g., Bimatoprost and Latanoprost) for the Treatment of Peri-Ocular Alopecia Areata

An UpToDate review on “Management of alopecia areata” (Messenger, 2017a) states that “Prostaglandin analogues -- Hypertrichosis of the eyelashes may occur as a side effect of glaucoma therapy with the topical prostaglandin analogues latanoprost and bimatoprost.  Knowledge of this effect led to clinical studies of the efficacy of these drugs for alopecia areata involving the eyelashes and eyebrows.  The majority of studies, including a 16-week randomized trial of 11 patients, have shown no benefit of these drugs for this disease.  However, a non-randomized, prospective study reported benefit with a longer course of therapy.  Of 44 patients with eyelash alopecia treated with latanoprost ophthalmic solution for 2 years, complete or moderate regrowth occurred in 17.5 and 27.5 %, respectively.  None of the 10 patients who did not receive the drug attained similar levels of response.  Additional studies are necessary to determine whether prostaglandin analogues should be utilized for the treatment of periocular alopecia areata”.

Ricar and colleagues (2022) noted that bimatoprost is a synthetic prostaglandin structural analog used among other indications to increase eyelash growth.  In a prospective, open-label study, these researchers examined the safety and effectiveness of topical bimatoprost in the treatment of eyelash loss in alopecia areata totalis (AT) and universalis (AU).  Subjects used ophthalmic bimatoprost (0.3 mg/ml) solution to the eyelid margins once-nightly for at least 12 weeks (mean treatment period was 30.6 weeks).  A total of 16 out of 17 subjects completed the study.  Only the subjects with eyelashes present at baseline experienced an increase in eyelash length and thickness.  No new eyelash regrowth was induced.  The authors concluded that in patients with AT and AU topical bimatoprost affected existing eyelashes; but failed to induce regrowth of new eyelashes.

Protein Tyrosine Phosphatase, Non-Receptor Type 22 (PTPN22) Gene Polymorphisms Testing

Salinas-Santander et al (2015) stated that the gene encoding the protein tyrosine phosphatase, non-receptor type 22 (PTPN22), which is exclusively expressed in immune cells, has been considered as a risk factor associated with a number of autoimmune diseases. In AA, the SNP, rs2476601, has been identified as a risk factor in several populations.  These researchers investigated the effect of PTPN22 C1858T inherited genetic polymorphism on the predisposition to severe forms of AA, in a case-control study on individuals.  The study included 64 unrelated patients diagnosed with several types of AA, as well as 225 healthy unrelated subjects.  The DNA samples were genotyped for PTPN22 C1858T polymorphism using PCR-restriction fragment length polymorphism technique.  Causal associations were determined by χ2 test and their respective OR was assessed in a 2×2 contingency table.  The results demonstrated a significant association of the T allele [p = 0.040; OR = 3.196; 95 % CI: 0.094 to 10.279] and the CT genotype (p = 0.038; OR = 3.313; 95 % CI: 1.008 to 10.892) with patchy AA.  The authors concluded that the findings of this study suggested the possible involvement of the T allele of the PTPN22 C1858T SNP as a genetic risk factor for this type of AA in the population studied.  These preliminary findings need to be validated by further research.

Simvastatin / Ezetimibe

In a prospective open study, Cho and colleagues (2017) examined the efficacy of the simvastatin/ezetimibe combination therapy for recalcitrant AA and investigated the relationship between various treatment responses and prognostic factors.  This trial was performed in patients with recalcitrant AA with the bald surface exceeding 75 %.  All patients took simvastatin (40 mg) and ezetimibe (10 mg) daily.  The extent of hair regrowth expressed as percentage of the bald area was used to evaluate the effectiveness of the therapy.  Of 14 enrolled patients, 4 patients (28.6 %) were judged as responders showing regrowth of 30 % to 80 % after 3 months of treatment.  The mean age of onset in non-responders was significantly lower than in responders.  The total score of prognostic factors, calculated as a sum of factors related to poor prognosis, was much lower in responders than in non-responders.  The authors concluded that the remission rate in this study was unsatisfactory.  However, since the recruited patients had not responded to any other treatments for AA, simvastatin/ezetimibe can still be considered as an alternative treatment for recalcitrant AA.  The total scores of the prognostic factors were statistically different between responders and non-responders.  These results can be used to predict the outcome of treatment with simvastatin/ezetimibe and anticipate prognosis.  Moreover, they stated that these findings need to be validated by future randomized placebo-controlled clinical trials in larger cohorts with precisely defined demographic features of patients.

The authors stated that the drawbacks of this study were the small number of patients (n = 14), relatively short follow-up period (3 months), and variations in treatment regimens before the start of the simvastatin/ezetimibe combination therapy.

In a prospective, observational study, Freitas Gouveia and Trueb (2017) examined the efficacy and tolerability of simvastatin/ezetimibe 40/10 mg over a treatment period of 6 months in AT, AU, multi-patch involvement of the scalp greater than 30 %, ophiasis, or diffuse AA.  Of the 12 patients included in the study, 67 % had no hair regrowth, 24 % transient diffuse or patchy hair regrowth, and 24 % patchy regrowth of pigmented hair that was not considered cosmetically satisfactory.  Adverse effects were observed in 24 % of patients, who reported myalgia; 1 patient showed elevation of creatine phosphokinase.  The authors concluded that simvastatin/ezetimibe was not effective for treatment of AA, at least in severe and/or cases refractory to other treatments, either as monotherapy or as adjuvant.

Topical Calcipotriol

In a retrospective, 12-week clinical trial, Cerman et al (2015) evaluated the safety and effectiveness of topical calcipotriol for the treatment of mild-to-moderate patchy AA. A total of 48 patients with mild-to-moderate AA were enrolled in this study.  Calcipotriol cream was applied to the affected areas twice-daily.  Severity of Alopecia Tool (SALT) score and hair regrowth rate were calculated at baseline and at 3, 6, 9, and 12 weeks.  At week 12, the total response was achieved in 69.2 % of patients.  When the mean SALT score of patients at week 12 was compared to that of patients at baseline, the value at week 12 was significantly lower (p= 0.001).  A regrowth score (RGS) greater than or equal to 3 (hair regrowth of greater than or equal to 50 %) was observed in 75 % of patients, whereas a RGS greater than or equal to 4 (hair regrowth of greater than or equal to 75 %) was observed in 62.5 % of patients and the complete regrowth rate (hair regrowth = 100 %) was 27.1 %.  The authors concluded that calcipotriol may serve as a safe and effective therapeutic option in mild-to-moderate patchy AA, and calls for more extensive controlled studies with this treatment.

Transporter 1 ATP-Binding Cassette Sub-Family B (MDR/TAP) Gene (TAP1) Gene Polymorphisms Testing

Kim et al (2015b) noted that the transporter 1 ATP-binding cassette sub-family B (MDR/TAP) gene (TAP1) is located in the major histocompatibility complex class II region, and forms a heterodimer that plays a key role in endogenous antigen presentation pathways. Investigation of polymorphisms identified in these loci has revealed an association with several autoimmune disorders. Alopecia areata is a common autoimmune disease resulting from T cell-induced damage to hair follicles. These investigators documented for the first time a comparison between the allelic and genotypic frequencies of TAP1 SNPs in patients with AA and those of a control group, using a direct sequencing method. The authors concluded that the findings of this study suggested an association between a promoter SNP (rs2071480) and susceptibility to this disease. These preliminary findings need to be validated by further research.

Unconventional Therapies for Alopecia Areata

Atanaskova Mesinkovska (2018) stated that in an attempt to better control their disease, patients with AA often seek alternative and somewhat unconventional therapies.  The internet search engine results yielded a plethora of promising products, many obtained without medical advice, whereas others require prescription or a visit with a physician.  Alternative treatments that have recently gained popularity among patients with AA include anti-histamines, cryotherapy, and low-dose naltrexone (LDN).  These unconventional AA remedies pose a challenge for patients and physicians alike, as they are not part of the standard AA therapeutic repertoire.  In addition, there is inadequate data evaluating their efficacy and even safety in AA.  Available evidence about the mechanisms of these therapeutic options pointed to a potential, but unproven, role in AA.

  • The cryotherapy technique used in the treatment of AA is not well described in the literature.  In most articles, it appeared that the entire area of hair loss is treated with a light spray jet every 2 weeks.
  • Naltrexone is an opiate antagonist used to treat addiction to heroin, morphine, and alcohol, typically at doses of 50 to 300 mg daily.  LDN at doses of 1 to 4.5 mg daily emerged as an anti-inflammatory treatment in the 1980s.  Since then, it has been evaluated in several small studies for the treatment of inflammatory conditions.  LDN is thought to work through modulation of inflammatory mediators and upregulation of endogenous opioid receptors.  Opioid peptides affect immune cytokine and chemokine signaling, and can function as immunomodulatory molecules as they have the ability to regulate T lymphocyte proliferation and block release of pro-inflammatory cytokines: IL-6 and IL-12, tumor necrosis factor-α, and nuclear factor NF-κB.  With regard to any role of LDN in the treatment of alopecia, there is not a single study describing its use in AA.  The only studies in alopecia were performed in trichotillomania, where LDN-treated patients did not have any observable differences in hair loss.  Although the research body on LDN in alopecia is minimal, both patients and prescribers are using it.  Because the presumed mechanism is anti-inflammatory, LDN can be potentially useful in inflammatory stages of AA and other alopecias.  It is important to note that naltrexone, even at low dose, may hyper-sensitize patients to exogenous opioids.  Prescribing physicians should carefully screen for potential drug interaction in patients on concurrent pain medications.

The authors concluded that the information on emerging unconventional therapies for AA should be interpreted and utilized with a sense of caution. Clinical studies are needed to better understand their mechanisms and potential role for AA.

Laser and Light Therapy Combined with Topical Minoxidil

In a systematic review and meta-analysis, Zhang et al (2023) examined the safety and effectiveness of laser and light therapy combined with topical minoxidil for the treatment of AA.  These investigators searched PubMed, Embase, Web of Science, the Cochrane Library, China National Knowledge Infrastructure (CNKI), China Biomedical Literature Database (CBM), VIP database, and Wanfang Data from their inception to September 18, 2022.  The risk of bias of the included RCTs was assessed by the Cochrane Collaboration tool.  RevMan 5.3 software and Stata 14.0 software were used to perform the statistical analysis.  The GRADE approach examined the quality of evidence; a total of 10 studies were included in this analysis.  The results of the meta-analysis showed that compared with topical minoxidil alone, the 308-nm excimer laser/light or He-Ne laser combined with topical minoxidil could reduce the SALT score (MD = -5.88, 95 % CI: -9.79 to -1.98, p = 0.003).  Whether fractional CO2 laser (RR = 1.29, 95 % CI: 1.14 to 1.46, p < 0.0001), 308-nm excimer laser/light (RR = 1.32, 95 % CI: 1.12 to 1.55, p = 0.001), He-Ne laser (RR = 1.69, 95 % CI: 1.07 to 2.69, p = 0.03), or NB-UVB (RR = 1.35, 95 % CI: 1.07 to 1.70, p = 0.01) combined with topical minoxidil may improve the treatment response rate, comparing with topical minoxidil only.  The recurrence rate of laser and light combined with topical minoxidil was lower than that of the minoxidil alone group (RR = 0.54, 95 % CI: 0.31 to 0.93, p = 0.03) when follow-up time was 1 year.  Furthermore, the incidence of AEs including irritant contact dermatitis, erythema, desquamation, pain, and pruritus was not significantly different between the 2 groups (RR = 1.50, 95 % CI: 0.95 to 2.36, p = 0.08).  The level of evidence for outcomes was classified as very low to moderate.  The authors concluded that based on the available evidence, laser and light combined with topical minoxidil therapy may be safe and effective for AA; however, more high-quality studies are needed for comprehensive analysis and further verification.

Psychological Interventions

Maloh et al (2023) noted that alopecia is associated with significant psychological burden.  There is limited evidence on the use of psychological interventions in conditions of hair loss.  In a systematic review, these investigators examined the available evidence on psychological treatments for QOL, mental health, and hair growth in various forms of alopecia.  PubMed and Embase were searched with pre-defined inclusion and exclusion criteria.  Reference lists were also examined for relevant studies.  A total of 9 studies met criteria and were included in this review; 8 of the studies were related to AA and 1 was related to scarring alopecia.  Mindfulness-based stress reduction (MBSR) was found to improve QOL-related subjective symptoms, relationship impacts, anxiety, phobia, distress, and psychological symptom intensity.  Alopecia-specific collocated behavioral health (CLBH) treatment showed a trend for psychosocial improvement in areas such as appearance shame, activity avoidance, negative emotions, and coping.  Hypnotherapy was found to improve anxiety and depression, QOL measures, and alexithymia.  There was also some evidence for significant hair growth with hypnosis; however, the data were mixed.  Psychotherapy combined with immunotherapy led to more hair growth; and supported self-confidence.  In addition, coping strategies modulated the subjective burden of alopecia, and were associated with disease improvement.  The authors concluded that further investigation is needed to better establish the effectiveness and optimal administration of these interventions in AA and scarring alopecia.


References

The above policy is based on the following references:

  1. Acikgoz G, Ozmen I, Cayirli M, et al. Pulse methylprednisolone therapy for the treatment of extensive alopecia areata. J Dermatolog Treat. 2014;25(2):164-166.
  2. Aghaei S. Topical immunotherapy of severe alopecia areata with diphenylcyclopropenone (DPCP): Experience in an Iranian population. BMC Dermatol. 2005;5:6.
  3. Al-Mutairi N. 308-nm excimer laser for the treatment of alopecia areata. Dermatol Surg. 2007;33(12):1483-1487.
  4. Alabdulkareem AS, Abahussein AA, Okoro A. Severe alopecia areata treated with systemic corticosteroids. Int J Dermatol. 1998;37(8):622-624.
  5. Ali S, Ibrahim AM, El Sayed N. Candida antigen immunotherapy versus steroid in the treatment of alopecia areata. Dermatol Ther. 2021;34(2):e14802.
  6. Alkhalifah A, Alsantali A, Wang E, et al. Alopecia areata update: Part II. Treatment. J Am Acad Dermatol. 2010;62(2):191-202, quiz 203-204.
  7. American Academy of Dermatology Committee on Guidelines of Care. Guidelines of care for phototherapy and photochemotherapy. J Am Acad Dermatol. 1994;31(4):643-648.
  8. Atanaskova Mesinkovska N. Emerging unconventional therapies for alopecia areata. J Investig Dermatol Symp Proc. 2018;19(1):S32-S33.
  9. Ayatollahi A, Hosseini H, Gholami J, et al. Platelet rich plasma for treatment of nonscarring hair loss: Systematic review of literature. J Dermatolog Treat. 2017:1-23.
  10. Aytekin N, Akcali C, Pehlivan S, et al. Investigation of interleukin-12, interleukin-17 and interleukin-23 receptor gene polymorphisms in alopecia areata. J Int Med Res. 2015;43(4):526-534.
  11. Bakry OA, El Farargy SM, El Shafiee MK, Soliman A. Serum vitamin D in patients with alopecia areata. Indian Dermatol Online J. 2016;7(5):371-377.
  12. Barton VR, Toussi A, Awasthi S, Kiuru M. Treatment of pediatric alopecia areata: A systematic review. J Am Acad Dermatol. 2022;86(6):1318-1334.
  13. Bayramgürler D, Demirsoy EO, Aktürk AŞ, Kıran R. Narrowband ultraviolet B phototherapy for alopecia areata. Photodermatol Photoimmunol Photomed. 2011;27(6):325-327.
  14. Behrens-Williams SC, Leiter U, Schiener R, et al. The PUVA-turban as a new option of applying a dilute psoralen solution selectively to the scalp of patients with alopecia areata. J Am Acad Dermatol. 2001;44(2):248-252.
  15. Bin Saif GA, Al-Khawajah MM, Al-Otaibi HM, et al. Efficacy and safety of oral mega pulse methylprednisolone for severe therapy resistant Alopecia areata. Saudi Med J. 2012;33(3):284-291.
  16. Bolduc C, Bissonnette R. Safety and efficacy of adalimumab for the treatment of severe alopecia areata: Case series of three patients. J Cutan Med Surg. 2012;16(4):257-260.
  17. Buckley DA, Du Vivier AW. The therapeutic use of topical contact sensitizers in benign dermatoses. Br J Dermatol. 2001;145(3):385-405.
  18. Byun JW, Moon JH, Bang CY, et al. Effectiveness of 308-nm excimer laser therapy in treating alopecia areata, determined by examining the treated sides of selected alopecic patches. Dermatology. 2015;231(1):70-76.
  19. Celik SD, Ates O. Genetic analysis of interleukin 18 gene polymorphisms in alopecia areata. J Clin Lab Anal. 2018;32(5):e22386.
  20. Cerman AA, Solak SS, Altunay İ, Kucukunal NA. Topical calcipotriol therapy for mild-to-moderate alopecia areata: A retrospective study. J Drugs Dermatol. 2015;14(6):616-620.
  21. Cervantes J, Perper M, Wong LL, et al. Effectiveness of platelet-rich plasma for androgenetic alopecia: A review of the literature. Skin Appendage Disord. 2018;4(1):1-11.
  22. Cho HR, Lew BL, Lew H, Sim WY. Treatment effects of intradermal botulinum toxin type A injection on alopecia areata. Dermatol Surg. 2010;36 Suppl 4:2175-2181.
  23. Choi JW, Suh DW, Lew BL, Sim WY. Simvastatin/ezetimibe therapy for recalcitrant alopecia areata: An open prospective study of 14 patients. Ann Dermatol. 2017;29(6):755-760.
  24. Coronel-Pérez IM, Rodríguez-Rey EM, Camacho-Martínez FM. Latanoprost in the treatment of eyelash alopecia in alopecia areata universalis. J Eur Acad Dermatol Venereol. 2010;24(4):481-485.
  25. Cotellessa C, Peris K, Caracciolo E, et al. The use of topical diphenylcyclopropenone for the treatment of extensive alopecia areata. J Am Acad Dermatol. 2001;44(1):73-76.
  26. Craiglow BG, Liu LY, King BA. Tofacitinib for the treatment of alopecia areata and variants in adolescents. J Am Acad Dermatol. 2017;76(1):29-32.
  27. Crowley EL, Fine SC, Katipunan KK, Gooderham MJ. The use of janus kinase inhibitors in alopecia areata: A review of the literature. J Cutan Med Surg. 2019;23(3):289-297.
  28. Cruciani M, Masiello F, Pati I, et al. Platelet-rich plasma for the treatment of alopecia: A systematic review and meta-analysis. Blood Transfus. 2023;21(1):24-36.
  29. Czarnecka A, Odziomek A, Murzyn M, et al. Wharton's jelly-derived mesenchymal stem cells in the treatment of four patients with alopecia areata. Adv Clin Exp Med. 2021;30(2):211-218.
  30. Dall'oglio F, Nasca MR, Musumeci ML, et al. Topical immunomodulator therapy with squaric acid dibutylester (SADBE) is effective treatment for severe alopecia areata (AA): Results of an open-label, paired-comparison, clinical trial. J Dermatolog Treat. 2005;16(1):10-14.
  31. de Oliveira AB, Alpalhao M, Filipe P, Maia-Silva J. The role of Janus kinase inhibitors in the treatment of alopecia areata: A systematic review. Dermatol Ther. 2019;32(5):e13053.
  32. Delamere FM, Sladden MM, Dobbins HM, Leonardi-Bee J. Interventions for alopecia areata. Cochrane Database Syst Rev. 2008;(2):CD004413.
  33. Dere G, Gundogdu M. Investigation of the relationship between alopecia areata and inflammatory blood parameters. J Cosmet Dermatol. 2021;20(12):4048-4051.
  34. Dillon KAL. A comprehensive literature review of JAK inhibitors in treatment of alopecia areata. Clin Cosmet Investig Dermatol. 2021;14:691-714.
  35. Doghaim NN, El-Tatawy RA, Neinaa YME, Abd El-Samd MM. Study of the efficacy of carboxytherapy in alopecia. J Cosmet Dermatol. 2018;17(6):1275-1285.
  36. Drake LA, Ceilley RI, Cornelison RL, et al. Guidelines of care for alopecia areata. J Am Acad Dermatol. 1992;26(2 Pt 1):247-250.
  37. Droitcourt C, Milpied B, Ezzedine K, et al. Interest of high-dose pulse corticosteroid therapy combined with methotrexate for severe alopecia areata: A retrospective case series. Dermatology. 2012;224(4):369-373.
  38. Elson DM. Hair disorders. In: Conn's Current Therapy. 51st ed. RE Rakel, ed. St. Louis, MO: W.B. Saunders Co.; 1999:794-797.
  39. Estebanez A, Estebanez N, Martín JM, Montesinos E. Apremilast in Refractory Alopecia Areata. Int J Trichology. 2019;11(5):213-215.
  40. Faghihi G, Andalib F, Asilian A. The efficacy of latanoprost in the treatment of alopecia areata of eyelashes and eyebrows. Eur J Dermatol. 2009;19(6):586-587.
  41. Farshi S, Mansouri P, Safar F, Khiabanloo SR. Could azathioprine be considered as a therapeutic alternative in the treatment of alopecia areata? A pilot study. Int J Dermatol. 2010;49(10):1188-1193.
  42. Freedberg IM, Eisen AZ, Wolff K, et al., eds. Dermatology in General Medicine. 5th ed. New York, NY: McGraw-Hill; 1999:737-739.
  43. Freire PCB, Petri V, Atallah AN. Minoxidil for patchy alopecia areata: Systematic review and meta-analysis. J Eur Acad Dermatol Venereol. 2019;33(9):1792-1799.
  44. Freitas Gouveia M, Trueb RM. Unsuccessful treatment of alopecia areata with simvastatin/ezetimibe: Experience in 12 patients. Skin Appendage Disord. 2017;3(3):156-160.
  45. Freyschmidt-Paul P, Happle R, McElwee KJ, Hoffmann R. Alopecia areata: Treatment of today and tomorrow. J Investig Dermatol Symp Proc. 2003;8(1):12-17.
  46. Freyschmidt-Paul P, Hoffmann R, Levine E, et al. Current and potential agents for the treatment of alopecia areata. Curr Pharm Des. 2001;7(3):213-230.
  47. Friedli A, Prins C, Chavaz P, et al. Unsuccessful treatment of alopecia areata universalis with extracorporeal photopheresis. Arch Dermatol. 2002;138(8):1103-1104.
  48. Fukumoto T, Fukumoto R, Magno E, et al. Treatments for alopecia areata: A systematic review and network meta-analysis. Dermatol Ther. 2021;34(3):e14916.
  49. Garg S, Messenger AG. Alopecia areata: Evidence-based treatments. Semin Cutan Med Surg. 2009;28(1):15-18.
  50. Georgala S, Katoulis AC, Befon A, et al. Inosiplex for treatment of alopecia areata: A randomized placebo-controlled study. Acta Derm Venereol. 2006;86(5):422-424.
  51. Gil-Quinones SR, Sepulveda-Pachon IT, Sanchez Vanegas G, Gutierrez-Castaneda LD. Effect of PTPN22, FAS/FASL, IL2RA and CTLA4 genetic polymorphisms on the risk of developing alopecia areata: A systematic review of the literature and meta-analysis. PLoS One. 2021;16(11):e0258499.
  52. Giordano S, Romeo M, di Summa P, et al. A meta-analysis on evidence of platelet-rich plasma for androgenetic alopecia. Int J Trichology. 2018;10(1):1-10.
  53. Gundogan C, Greve B, Raulin C. Treatment of alopecia areata with the 308-nm xenon chloride excimer laser: Case report of two successful treatments with the excimer laser. Lasers Surg Med. 2004;34(2):86-90.
  54. Guo L, Feng S, Sun B, et al. Benefit and risk profile of tofacitinib for the treatment of alopecia areata: A systemic review and meta-analysis. J Eur Acad Dermatol Venereol. 2020;34(1):192-201.
  55. Gupta AK, Carviel JL. Meta-analysis of 308-nm excimer laser therapy for alopecia areata. J Dermatolog Treat. 2021;32(5):526-529.
  56. Gupta AK, Carviel JL, Foley KA, et al. Monotherapy for alopecia areata: A systematic review and network meta-analysis. Skin Appendage Disord. 2019;5(6):331-337.
  57. Gupta V, Parihar AS, Sharma VK, et al. Evaluation of platelet-rich plasma on hair regrowth and lesional T-cell cytokine expression in alopecia areata: A randomized observer-blinded, placebo-controlled, split-head pilot study. J Am Acad Dermatol. 2021;84(5):1321-1328.
  58. Habif TB. Clinical Dermatology. 3rd ed. St Louis, MO: Mosby-Year Book; 1996:752-754.
  59. Hamdino M, El-Barbary RA, Darwish HM, et al. Intralesional methotrexate versus triamcinolone acetonide for localized alopecia areata treatment: A randomized clinical trial. J Cosmet Dermatol. 2022;21(2):707-715.
  60. Healy E, Rogers S. PUVA treatment for alopecia areata-does it work? A retrospective review of 102 cases. Br J Dermatol. 1993;129(1):42-44.
  61. Hordinsky M, Donati A. Alopecia areata: An evidence-based treatment update. Am J Clin Dermatol. 2014;15(3):231-246.
  62. Hull SM, Pepall L, Cunliffe WJ. Alopecia areata in children: Response to treatment with diphencyprone. Br J Dermatol. 1991;125(2):164-168.
  63. Hunter N, Shaker O, Marei N. Diphencyprone and topical tacrolimus as two topical immunotherapeutic modalities. Are they effective in the treatment of alopecia areata among Egyptian patients? A study using CD4, CD8 and MHC II as markers. J Dermatolog Treat. 2011;22(1):2-10.
  64. Iorizzo M, Tosti A. Emerging drugs for alopecia areata: JAK inhibitors. Expert Opin Emerg Drugs. 2018;23(1):77-81.
  65. Islamoglu ZGK, Demirbas A. Evaluation of complete blood cell and inflammatory parameters in patients with alopecia areata: Their association with disease severity. J Cosmet Dermatol. 2020;19(5):1239-1245.
  66. Ismail FF, Sinclair R. JAK inhibition in the treatment of alopecia areata -- a promising new dawn? Expert Rev Clin Pharmacol. 2020;13(1):43-51.
  67. Jabbari A, Dai Z, Xing L, et al. Reversal of alopecia areata following treatment with the JAK1/2 inhibitor baricitinib. EBioMedicine. 2015;2(4):351-355.
  68. Jabbari A, Sansaricq F, Cerise J, et al. An open-label pilot study to evaluate the efficacy of tofacitinib in moderate to severe patch type alopecia areata, totalis and universalis. J Invest Dermatol. 2018;138(7):1539-1545.
  69. Ji C, Liu S, Zhu K, et al. HLA-DRB1 polymorphisms and alopecia areata disease risk: A systematic review and meta-analysis. Medicine (Baltimore). 2018;97(32):e11790.
  70. Jin W, Zheng H, Shan B, Wu Y. Changes of serum trace elements level in patients with alopecia areata: A meta-analysis. J Dermatol. 2017;44(5):588-591.
  71. Kalkan G, Seckin HY, Benli İ, et al. Relationship between manganese superoxide dismutase (MnSODAla-9Val) and glutathione peroxidase (GPx1 Pro 197 Leu) gene polymorphisms and alopecia areata. Int J Clin Exp Med. 2015;8(11):21533-21540.
  72. Kim HK, Lee H, Lew BL, et al. Association between TAP1 gene polymorphisms and alopecia areata in a Korean population. Genet Mol Res. 2015b;14(4):18820-18827.
  73. Kim SK, Chung JH, Park HJ, et al. Polymorphisms in the promoter regions of the CXCL1 and CXCL2 genes contribute to increased risk of alopecia areata in the Korean population. Genet Mol Res. 2015a;14(3):9667-9674.
  74. Kose O, Safali M, Bulent Tastan H, Gur AR. Mycophenolate mofetil in extensive alopecia areata: No effect in seven patients. Dermatology. 2004;209(1):69-70.
  75. Kostovic K, Gulin SJ, Mokos ZB, Ceovic R. Tofacitinib, an oral janus kinase inhibitor: Perspectives in dermatology. Curr Med Chem. 2017;24(11):1158-1167.
  76. Lai VWY, Chen G, Gin D, Sinclair R. Systemic treatments for alopecia areata: A systematic review. Australas J Dermatol. 2019;60(1):e1-e13.
  77. Lamb RC, Young D, Holmes S. Retrospective review of diphencyprone in the treatment of alopecia areata. Clin Exp Dermatol. 2016;41(4):352-358.
  78. Lee HW, Jun JH, Lee JA, et al. Acupuncture for treating alopecia areata: A protocol of systematic review of randomised clinical trials. BMJ Open. 2015;5(10):e008841.
  79. Lee JH, Eun SH, Kim SH, et al. Excimer laser/light treatment of alopecia areata: A systematic review and meta-analyses. Photodermatol Photoimmunol Photomed. 2020;36(6):460-469.
  80. Lux-Battistelli C. Combination therapy with zinc gluconate and PUVA for alopecia areata totalis: An adjunctive but crucial role of zinc supplementation. Dermatol Ther. 2015;28(4):235-238.
  81. MacDonald Hull SP, Wood ML, Hutchinson PE, et al. Guidelines for the management of alopecia areata. Br J Dermatol. 2003;149(4):692-699.
  82. Madani S, Shapiro, J. Alopecia areata update. J Am Acad Dermatol. 2000;42(4):549-566.
  83. Majid I, Jeelani S, Imran S. Fractional carbon dioxide laser in combination with topical corticosteroid application in resistant alopecia areata: A case series. J Cutan Aesthet Surg. 2018;11(4):217-221.
  84. Maloh J, Engel T, Natarelli N, et al. Systematic review of psychological interventions for quality of life, mental health, and hair growth in alopecia areata and scarring alopecia. J Clin Med. 2023;12(3):964.
  85. Maria-Angeliki G, Alexandros-Efstratios K, Dimitris R, Konstantinos K. Platelet-rich plasma as a potential treatment for noncicatricial alopecias. Int J Trichology. 2015;7(2):54-63.
  86. Mehraban S, Feily A. 308nm excimer laser in dermatology. J Lasers Med Sci. 2014;5(1):8-12.
  87. Messenger AG. Alopecia areata: Management. UpToDate [online serial]. Waltham, MA: UpToDate; reviewed February 2022.
  88. Messenger AG, McKillop J, Farrant P, et al. British Association of Dermatologists' guidelines for the management of alopecia areata 2012. Br J Dermatol. 2012;166(5):916-926. 
  89. Messenger AG. Alopecia areata: Management. UpToDate [online serial]. Waltham, MA: UpToDate; reviewed February 2021.
  90. Messenger AG. Clinical manifestations and diagnosis of alopecia areata. UpToDate [online serial]. Waltham, MA: UpToDate; reviewed February 2017b.
  91. Messenger AG. Management of alopecia areata. UpToDate [online serial]. Waltham, MA: UpToDate; reviewed February 2017a.
  92. Mikhaylov D, Glickman JW, Del Duca E, et al. A phase 2a randomized vehicle-controlled multi-center study of the safety and efficacy of delgocitinib in subjects with moderate-to-severe alopecia areata. Arch Dermatol Res. 2023;315(2):181-189.
  93. Muscianese M, Magri F, Leoncini P, et al. Alopecia areata treated with topical and systemic brevilin A: A case series. Dermatol Ther. 2021;34(2):e14778.
  94. Nasiri S, Haghpanah V, Taheri E, et al. Hair regrowth with topical triiodothyronine ointment in patients with alopecia areata: A double-blind, randomized pilot clinical trial of efficacy. J Eur Acad Dermatol Venereol. 2012;26(5):654-656. 
  95. National Alopecia Areata Foundation (NAAF) [Web Site]. San Rafael, CA: NAAF; 2002. Available at: http://www.alopeciaareata.com/. Accessed January 13, 2003.
  96. Nouh AH, Kadah AS, Said M. Comparative study of the use of fractional CO 2 laser versus the use of liquid nitrogen cryotherapy in the treatment of alopecia areata in a sample of the Egyptian population. Dermatol Ther. 2022;35(4):e15358.
  97. Olsen EA, Kornacki D, Sun K, et al. Ruxolitinib cream for the treatment of patients with alopecia areata: A 2-part, double-blind, randomized, vehicle-controlled phase 2 study. J Am Acad Dermatol. 2020;82(2):412-419.
  98. Orecchia G, Malagoli P, Santagostino L. Treatment of severe alopecia areata with squaric acid dibutylester in pediatric patients. Pediar Dermatol. 1994;11(1):65-68.
  99. Park KY, Jang WS, Son IP, et al. Combination therapy with cyclosporine and psoralen plus ultraviolet a in the patients with severe alopecia areata: A retrospective study with a self-controlled design. Ann Dermatol. 2013;25(1):12-16.
  100. Parker F. Skin diseases of general importance. In: Cecil Textbook of Medicine. 20th ed. JC Bennett, F Plum, eds. Philadelphia, PA: W.B. Saunders Co.; 1996:2215-2217.
  101. Phan K, Sebaratnam DF. JAK inhibitors for alopecia areata: A systematic review and meta-analysis. J Eur Acad Dermatol Venereol. 2019;33(5):850-856.
  102. Price VH, Willey A, Chen BK. Topical tacrolimus in alopecia areata. J Am Acad Dermatol. 2005;52(1):138-139.
  103. Price VH. Treatment of hair loss. N Engl J Med. 1999;341(13):964-973.
  104. Ricar J, Cetkovska P, Hordinsky M, Ricarova R. Topical bimatoprost in the treatment of eyelash loss in alopecia totalis and universalis: A prospective, open-label study. Dermatol Ther. 2022;35(6):e15438.
  105. Salinas-Santander M, Sanchez-Domínguez C, Cantu-Salinas C, et al. Association between PTPN22 C1858T polymorphism and alopecia areata risk. Exp Ther Med. 2015;10(5):1953-1958.
  106. Salman A, Sarac G, Ergun T. Alopecia universalis unresponsive to treatment with tofacinitib: Report of a case with a brief review of the literature. Dermatol Online J. 2017;23(7).
  107. Samadi A, Ahmad Nasrollahi S, Hashemi A, et al. Janus kinase (JAK) inhibitors for the treatment of skin and hair disorders: A review of literature. J Dermatolog Treat. 2017;28(6):476-483.
  108. Sams WM Jr, Lynch PJ, eds. Principles and Practice of Dermatology. 2nd ed. New York, NY: Churchill Livingstone; 1996:783-787.
  109. Shapiro J, Tan J, Ho V, et al. Treatment of chronic severe alopecia areata with topical diphenylcyclopropenone and 5% minoxidil: A clinical and immunopathologic evaluation. J Am Acad Dermatol. 1993;29(5 Pt 1):729-735.
  110. Shapiro, J, Madani S. Alopecia areata: Diagnosis and management. Int J Dermatol. 1999;38 Suppl 1:19-24.
  111. Sharma VK, Gupta S. Twice weekly 5 mg dexamethasone oral pulse in the treatment of extensive alopecia areata. J Dermatol. 1999;26(9):562-565.
  112. Sharma VK. Pulsed administration of corticosteroids in the treatment of alopecia areata. Int J Dermatol. 1996;35(2):133-136.
  113. Shellow WR. Approach to the patient with hair loss. In: Primary Care Medicine. AH Goroll, LA May, AG Mulley, eds. Philadelphia, PA: J.B. Lippincott; 1995;182:899-902.
  114. Shreberk-Hassidim R, Ramot Y, Zlotogorski A. Janus kinase inhibitors in dermatology: A systematic review. J Am Acad Dermatol. 2017;76(4):745-753.
  115. Staumont-Salle D, Vonarx M, Lengrand F. Pulse corticosteroid therapy for alopecia areata: Long-term outcome after 10 years. Dermatology. 2012;225(1):81-87.
  116. Taylor CR, Hawk JL. PUVA treatment of alopecia areata partialis, totalis and universalis: Audit of 10 years' experience at St. John's Institute of Dermatology. Br J Dermatol. 1995;1333(6):914-918.
  117. Thiers BH. Topical tacrolimus: Treatment failure in a patient with alopecia areata. Arch Dermatol. 2000;136(1):124.
  118. Tizaoui K, Kim SH, Jeong GH, et al. Association of PTPN22 1858C/T polymorphism with autoimmune diseases: A systematic review and Bayesian approach. J Clin Med. 2019;8(3).
  119. Tkachenko E, Okhovat JP, Manjaly P, et al. Complementary & alternative medicine for alopecia areata: A systematic review. J Am Acad Dermatol. 2023;88(1):131-143.
  120. Trink A, Sorbellini E, Bezzola P, et al.  A randomized, double-blind, placebo- and active-controlled, half-head study to evaluate the effects of platelet-rich plasma on alopecia areata. Br J Dermatol. 2013;169(3):690-694.
  121. Triyangkulsri K, Suchonwanit P. Role of janus kinase inhibitors in the treatment of alopecia areata. Drug Des Devel Ther. 2018;12:2323-2335.
  122. Trost LB, Bergfeld WF, Calogeras E. The diagnosis and treatment of iron deficiency and its potential relationship to hair loss. J Am Acad Dermatol. 2006;54(5):824-844. 
  123. United States Pharmacopeial Convention, Inc. USP Dispensing Information. Vol I-Drug Information for the Health Care Professional. 19th ed. Englewood, CO: Micromedex, Inc.; 1999;1465-1468, 2021-2023.
  124. van den Biggelaar FJ, Smolders J, Jansen JF. Complementary and alternative medicine in alopecia areata. Am J Clin Dermatol. 2010;11(1):11-20.
  125. Van der Steen PH, Boezeman JB, Happle R. Topical immunotherapy for alopecia areata: Re-evaluation of 139 cases after an additional follow-up periods of 19 months. Dermatology. 1992;184(3):198-201.
  126. Vazquez OA, Safeek RH, Komberg J, Becker H. Alopecia areata treated with advanced platelet-rich fibrin using micronization. Plast Reconstr Surg Glob Open. 2022;10(1):e4032.
  127. Waldmann TA. The biology of IL-15: implications for cancer therapy and the treatment of autoimmune disorders. J Investig Dermatol Symp Proc. 2013;16(1):S28-S30.
  128. Wat H, Dytoc M. Off-label uses of topical vitamin d in dermatology: A systematic review. J Cutan Med Surg. 2014;18(2):91-108.
  129. Willemsen R, Vanderlinden J, Deconinck A, Roseeuw D. Hypnotherapeutic management of alopecia areata. J Am Acad Dermatol. 2006;55(2):233-237.
  130. Wojciechowska-Zdrojowy M, Jankowska-Konsur A, Nowicka-Suszko, et al. Comparison of serum concentrations of interleukins 10, 12, 17 and 35 between patients with alopecia areata and controls. Postepy Dermatol Alergol. 2021;38(6):1052-1057.
  131. Zaher H, Gawdat HI, Hegazy RA, Hassan M. Bimatoprost versus mometasone furoate in the treatment of scalp alopecia areata: A pilot study. Dermatology. 2015;230(4):308-313.
  132. Zakaria W, Passeron T, Ostovari N, et al. 308-nm excimer laser therapy in alopecia areata [letter]. J Am Acad Dermatol. 2004;51(5):837-838.
  133. Zarei M, Wikramanayake TC, Falto-Aizpurua L, et al. Low level laser therapy and hair regrowth: An evidence-based review. Lasers Med Sci. 2016;31(2):363-371.
  134. Zhang J, Lin P, Lin H, et al. Laser and light therapy combined with topical minoxidil for alopecia areata: A systematic review and meta-analysis of randomized controlled trials. Lasers Med Sci. 2023;38(1):74.