Next Article in Journal
Patient-Initiated Follow-Up in Ovarian Cancer
Next Article in Special Issue
Quality of Life in Cancer Patients: The Modern Psycho-Oncologic Approach for Romania—A Review
Previous Article in Journal
Assessing Psychological Morbidity in Cancer-Unaffected BRCA1/2 Pathogenic Variant Carriers: A Systematic Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Prevention and Treatment of Chemotherapy-Induced Alopecia: What Is Available and What Is Coming?

by
Tongyu C. Wikramanayake
*,
Nicole I. Haberland
,
Aysun Akhundlu
,
Andrea Laboy Nieves
and
Mariya Miteva
Dr. Phillip Frost Department of Dermatology & Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
*
Author to whom correspondence should be addressed.
Curr. Oncol. 2023, 30(4), 3609-3626; https://doi.org/10.3390/curroncol30040275
Submission received: 15 February 2023 / Revised: 21 March 2023 / Accepted: 22 March 2023 / Published: 25 March 2023
(This article belongs to the Special Issue Quality of Life and Side Effects Management in Cancer Treatment)

Abstract

:
Millions of new cancer patients receive chemotherapy each year. In addition to killing cancer cells, chemotherapy is likely to damage rapidly proliferating healthy cells, including the hair follicle keratinocytes. Chemotherapy causes substantial thinning or loss of hair, termed chemotherapy-induced alopecia (CIA), in approximately 65% of patients. CIA is often ranked as one of the most distressing adverse effects of chemotherapy, but interventional options have been limited. To date, only scalp cooling has been cleared by the US Food and Drug Administration (FDA) to prevent CIA. However, several factors, including the high costs not always covered by insurance, preclude its broader use. Here we review the current options for CIA prevention and treatment and discuss new approaches being tested. CIA interventions include scalp cooling systems (both non-portable and portable) and topical agents to prevent hair loss, versus topical and oral minoxidil, photobiomodulation therapy (PBMT), and platelet-rich plasma (PRP) injections, among others, to stimulate hair regrowth after hair loss. Evidence-based studies are needed to develop and validate methods to prevent hair loss and/or accelerate hair regrowth in cancer patients receiving chemotherapy, which could significantly improve cancer patients’ quality of life and may help improve compliance and consequently the outcome of cancer treatment.

Graphical Abstract

1. Introduction

Globally, 18.1 million people were diagnosed with cancer (excluding non-melanoma skin cancers) in 2020, and this number is projected to rise to 26 million in 2040 [1,2]. By 2040, the number of patients requiring first-course chemotherapy each year will increase by approximately 50% to 15.0 million [2]. Among patients receiving chemotherapy, an estimated 65% are expected to develop chemotherapy-induced alopecia (CIA) and possible changes in hair pigmentation, texture, quantity, and growth [3,4]. While many common targeted therapies and some immunotherapies are classified as chemotherapy in the National Cancer Database (NCDB), in this review we focus on CIA induced by cytotoxic drugs [5].
CIA is one of the most visible and dreaded adverse effects of systemic cytotoxic drugs for both male and female patients [4,6,7]. Cytotoxic drugs kill rapidly proliferating cells, not only the cancer cells but also highly proliferative healthy cells such as hematopoietic cells, intestinal epithelial cells, and hair follicle (HF) keratinocytes during the growth phase called anagen. Within the anagen HF, the main targets of cytotoxic drugs are the highly proliferative keratinocytes in the hair matrix at the bottom of the HF and their pigmentary system, leading to rapid apoptosis and hair shaft breakage/shedding [8]. Because up to 90% of scalp HFs are in the growth phase at any given time, scalp hair loss is often massive within weeks of the initiation of chemotherapy [9]. The risk of CIA and the degree of hair loss differ substantially based on the medication, dose, frequency, duration, and route of administration. The incidence of CIA is more than 80% for antimicrotubule agents (e.g., paclitaxel), 60–100% for topoisomerase inhibitors (e.g., doxorubicin), more than 60% for alkylators (e.g., cyclophosphamide), and 10–50% for antimetabolites (e.g., 5-fluorouracil and leucovorin) [10]. Combination therapy can further increase the incidence. Beard, eyebrow, and eyelashes can also be affected, and the severity of these hair defects may vary by the specific drugs and treatment regimens [4,7]. While alopecia is generally reversible 3–6 months after cessation of chemotherapy (the new hair may show different texture and color), some patients experience no hair regrowth or only partial hair regrowth. Permanent CIA (pCIA) is diagnosed when there is no hair regrowth or incomplete hair regrowth 6 months after chemotherapy cessation. The incidence of pCIA is dose- and schedule-related and has been associated with docetaxel given at doses of 75 mg/m2 or higher per cycle, and less commonly paclitaxel [11,12,13,14]. In a study of 383 patients treated for breast cancer in the UK, pCIA was reported by 23.3% of patients receiving docetaxel (among a total of 245 patients) and 10.1% paclitaxel (among a total of 138 patients) (p < 0.01) [13]. In a study of 61 patients treated for breast cancer in Korea, the incidences of pCIA at 6 months and 3 years were 39.5% and 42.3%, respectively [12]. As a result, pCIA (together with vision damage) is at the center of thousands of lawsuits against docetaxel (sold under Taxotere) manufacturer Sanofi-Aventis. Depletion of HF keratinocyte stem cells via apoptosis, DNA damage, and epithelial-mesenchymal transition were detected upon chemotherapy in human scalp HF organ culture and may be one of the underlying mechanisms of pCIA [15,16].
Given that scalp and facial hair are key elements of good health, beauty, and youth in social communication [17], CIA has significant negative impact on patients’ self-esteem, body image, sexuality, and overall quality of life [4,7,8,18]. In a study of 179 male and female patients who developed CIA, 101 (56.4%) patients felt that alopecia was the worst side effect of chemotherapy, 129 (72%) patients said hair loss was affecting their social life, and 37 (20.6%) patients used hair accessories to hide the hair loss [6]. Cancer patients would even consider rejecting life-saving chemotherapy for fear of CIA [4,19]. Being a public sign of the illness, the visibility of alopecia makes it difficult for patients to keep their cancer status private; the moment alopecia becomes apparent is often the moment of public recognition of the patient’s cancer treatment [20]. Consequently, patients with CIA may begin to perceive certain changes in the attitudes towards them, ranging from sympathy to rejection [20]. There are excellent recent reviews on the clinical features, diagnosis, pathobiology of CIA, and CIA effects on quality of life [3,4,9,21,22,23,24].
Despite the severe negative impact of CIA on a large number of patients, preventative and treatment options have been limited for CIA. To date, only one approach, i.e., scalp cooling, has been cleared by the US Food and Drug Administration (FDA) in 2015 to prevent CIA. In recent years, topical and oral minoxidil, photobiomodulation therapy (PBMT), platelet-rich plasma (PRP) injections, and other oral and topical treatments that have shown efficacy in promoting hair growth in other alopecia disorders are explored to stimulate hair regrowth after CIA. Here we review the current state of CIA prevention and treatment and discuss new approaches that are being tested in clinical trials. Given the negative psychological and emotional impact of CIA, approaches that successfully prevent hair loss or accelerate hair regrowth will significantly improve patients’ quality of life and may help improve compliance and consequently the outcome of cancer treatment.

2. CIA Prevention: Scalp Cooling and Topical Vasoconstrictors

2.1. Scalp Cooling Systems and Cold Caps

Scalp cooling has been used since the late 1970s to prevent CIA, and the scalp cooling system DigniCap was the first device cleared by the U.S. Food and Drug Administration (FDA) for the prevention of CIA in 2015 in breast cancer patients [9]. Scalp cooling prevents or reduces CIA via two main mechanisms: first, by reducing the scalp temperature using cold air, gel packs, or electronically cooled caps, local vasoconstriction reduces drug inflow to the scalp, consequently limiting the local concentration of chemotherapeutic agents in the HFs; second, the low temperature also helps to reduce cellular metabolism in the HFs, making them less vulnerable to the antimitotic and antimetabolic effects of chemotherapeutic drugs [20]. Additionally, low temperature-induced cell cycle arrest at the G0/G1 phase, increased HSP70 accumulation to protect cells from stresses, and reduced cell apoptosis have also been shown as potential mechanisms of HF protection against CIA [25].
Currently, there are several FDA-cleared scalp cooling systems available to patients, as well as cold caps that are not yet cleared by the FDA (Table 1). The cooling systems are mobile cooling units that pump circulating cold coolant to cool the scalp, with the temperature accurately controlled by a computer. They are operated by healthcare providers (Table 1). The cold caps use 3–8 caps containing coolant gels that can be re-frozen quickly. Temperature is carefully monitored, and the caps are changed manually. They are self-administered by the patient’s companion (Table 1). While some costs may be reimbursed by some insurance plans for the FDA-cleared cooling systems, others are not, and costs and access remain an important issue for many patients.
To achieve effective protection, the scalp must attain a subcutaneous (1–2 mm) temperature below 22 °C, which is equivalent to an epicutaneous temperature of 19 °C, although greater preventive effects could be achieved with temperatures close to 15 °C [20,26]. Scalp cooling begins approximately 30 min before chemotherapy starts, continues during the infusion, and must continue for a set period after the conclusion of treatment. The post-infusion cooling time depends on the pharmacokinetics of the chemotherapeutic agents and doses used, typically 60–180 min. The cooling cap remains on the scalp for another 5–10 min to return to room temperature. Adverse effects such as a feeling of coldness, neck and shoulder discomfort, headache, scalp pain, forehead pain, nausea, and/or light-headedness and dizziness have been reported, but are temporary [27]. However, patient compliance is high, and efficacy of protection from hair loss by scalp cooling has been increasingly demonstrated over the past 20 years, with average success rates of 50–70% [27]. It is important to note that efficacy for hair preservation depends on chemotherapy type, e.g., higher efficacy is associated with a taxane-alone regimen, and much lower efficacy is associated with an anthracycline-based regimen [28]. Over 70% of current clinical trials aimed at prevention and treatment of CIA are testing scalp cooling devices, alone or in combination with other approaches (Table 2).
While the FDA has cleared the expanded use of scalp cooling systems to patients with solid tumors undergoing chemotherapeutic protocols associated with a high risk of developing CIA, scalp cooling is not applicable to all chemotherapy regimens. For example, patients receiving platinum derivatives who develop severe peripheral neuropathies, which limit their tolerance to cold, should avoid scalp cooling [29]. Patients with hematological tumors should also avoid scalp cooling for their increased risk of scalp metastasis [9]. Furthermore, scalp cooling devices should be avoided for patients with cold agglutinin disease, cryoglobulinemia, and posttraumatic cold injury due to the risk of triggering local or generalized attack [30].

2.2. Topical Calcitriol

Vitamin D, partly synthesized within the keratinocytes in the presence of ultraviolet-B radiation and partly obtained from diet, plays an important role in dermatology and dermatotherapeutics due to its anti-inflammatory and immunomodulatory properties, as well as regulation of keratinocyte differentiation and proliferation [31]. Vitamin D is also believed to stimulate hair growth and anagen initiation, and its deficiency has been closely linked to various types of alopecia, including telogen effluvium, androgenetic alopecia, alopecia areata, trichotillomania, and scarring alopecia [31,32]. Although previous studies have failed to detect positive effects of calcitriol on patients with CIA—in fact, application of calcitriol (1,25-dihydroxyvitamin D3) caused a pruritic irritative dermatitis in five of the eight patients—further investigation of the drug was suggested because patients did not experience any systemic toxic effects [33]. Topical calcitriol was applied to 23 female breast and gynecologic cancer patients who were receiving a taxane-based chemotherapy, and a reduction of alopecia of >50% was observed in 8 patients at Week 7 (NCT01588522) (Table 2) [34]. The safe dosage was determined as 80 μg/mL [34].

2.3. Keratinocyte Growth Factor (KGF)

Keratinocyte growth factor (KGF), also called fibroblast growth factor 7 (FGF7), is a potent mitogen that regulates the migration and differentiation of various epithelial cells and protects them from various insults under stress conditions [35]. Intradermal injections of a bioengineered hair formulation containing various growth factors, including KGF, into the scalp, once every 3 weeks for a total of eight sessions, resulted in significant reduction in hair fall, decreased number of vellus hairs, and increased number of terminal hairs and shaft diameter [36]. In addition, in a neonatal rat model of cytosine arabinoside-induced alopecia, pretreatment with recombinant KGF alone induced a dose-dependent cytoprotective effect, abrogating as much as 50% of the alopecia [37]. Furthermore, in human scalp HF organ culture, KGF pretreatment slightly, but significantly, inhibited HF apoptosis and dystrophy induced by 4-hydroperoxycyclophosphamide (4-HC), a key cyclophosphamide metabolite [38]. A study to investigate KGF hair serum for the prevention of CIA (NCT04554732) was recently completed (Table 2).

2.4. Topical Vasoconstrictors

Topical vasoconstrictors have also been shown to be effective in protecting against CIA and oral mucositis, but only in animal models to date. In one study, when epinephrine was topically applied to the dorsal skin of neonatal rats, a 95% coat retention (suppression of alopecia) was observed in rats treated with N-nitroso-N-methylurea, and a 16% coat retention was observed in rats treated with systemic Cytoxan [39]. The promising result justifies further testing to determine whether vasoconstrictors can protect against CIA in humans. A major advantage of vasoconstrictors over scalp cooling is that they can be applied when necessary, while scalp cooling is only administered at the time of infusion. Because the half-life of chemotherapeutic drugs is longer than their infusion time, vasoconstriction through scalp cooling at the time of infusion cannot prevent their toxic effects in the subsequent days/weeks. Additionally, topical vasoconstrictors can be used for patients who are not good candidates for scalp cooling [9].

2.5. Low Intensity Ultrasound (LIUS)

Recently, low intensity ultrasound (LIUS) was shown to reverse the cytotoxic damage of paclitaxel on microtubules in cultured cells [40,41]. Given their excellent safety profile and availability as inexpensive home devices, it is important to explore the potential of LIUS application to prevent CIA.

3. CIA Treatment

It often takes several months for hair to grow back after CIA. Several agents, physical methods, and injections that have shown efficacy in promoting hair regrowth in other types of alopecia—such as alopecia areata and androgenetic alopecia—have been explored for their ability to expedite hair regrowth after CIA. These include topical and oral minoxidil, bimatoprost/prostaglandin analog, photobiomodulation therapy (PBMT), platelet-rich plasma (PRP), spironolactone, and other agents, as well as complementary and alternative treatments. These agents, physical methods, and injections are discussed below.

3.1. Minoxidil

Minoxidil is a pyrimidine N-oxide that acts as an antihypertensive vasodilator [42]. It induces angiogenesis by upregulating vascular endothelial growth factor expression and activating prostaglandin endoperoxide synthase 1, which can stimulate hair growth [43]. Moreover, minoxidil alters the temporal aspects of the hair cycle by extending the HF growth phase—termed anagen—while shortening the relative quiescent phase—termed telogen [9,44]. Minoxidil was approved by the FDA to treat male and female pattern hair loss in 1988, and 2% or 5% topical minoxidil solution or 5% foam has been widely used to promote hair regrowth, including off-label use for other types of alopecia, including CIA [9,44,45,46]. In a double-blind randomized controlled study, women who were administered topical minoxidil 2% twice a day 4 months post-chemotherapy showed a statistically significant decrease (p = 0.03) for the alopecic period, reducing it from 137 days for the placebo to 87 days [21,47]. Additionally, a recent controlled study found topical minoxidil to be effective in promoting hair regrowth after taxane-based chemotherapy, even in the case of pCIA [11]. Furthermore, oral minoxidil has also shown efficacy. A low dosage of oral minoxidil, starting at 1.25 mg daily, has also been used in the treatment of pCIA [48]. Oral minoxidil treatment would be particularly useful for patients with poor compliance to topical minoxidil due to scalp irritation or hair texture. A review that looked at a total of 17 studies with 634 patients using oral minoxidil as a primary treatment for hair loss (mostly AGA but including CIA) found that it was an effective alternative treatment for patients who had trouble using the topical minoxidil solution [46].
However, because vasodilation leads to a greater permanence of anticancer drug around the HF, minoxidil should not be used during chemotherapy but should be administered after chemotherapy discontinuation to achieve a greater regrowth rate [9]. To determine the optimal time to administer topical minoxidil, the pharmacokinetics of the chemotherapeutic drug needs to be considered, and a topical hydrocortisone should also be administered with minoxidil to act as an anti-inflammatory agent and support hair growth [9,44,47]. Interestingly, in murine studies, minoxidil not only significantly improved the hair quality (both length and growth) after paclitaxel treatment, but also suppressed neuroinflammation, as well as showing a synergistic anti-tumor effect with paclitaxel in tumor xenograft models of both cervical and breast cancer [49]. Therefore, minoxidil is an excellent option for treating CIA, as well as a potential candidate to prevent chemotherapy-induced peripheral neuropathy. There is an ongoing clinical trial (NCT03831334) evaluating the effect of low-dose oral minoxidil as treatment for pCIA (Table 2).

3.2. Bimatoprost/Prostaglandin F2α Analog

Bimatoprost is a synthetic analog of prostaglandin F2α. It increases the outflow of aqueous fluid from the eye and lowers intraocular pressure, and it has been used to treat conditions with high pressure inside the eye, including glaucoma [50,51]. Because the association of acquired trichomegaly of the eyelashes and hypertrichosis was discovered with bimatoprost use [52,53], bimatoprost (0.03%) has been used to treat eyelash hypotrichosis and eyelash, eyebrow, and scalp alopecia [54,55], as well as for aesthetics purposes. Several controlled clinical studies have shown the efficacy of bimatoprost gel for chemotherapy-induced ciliary hypotrichosis, reporting a faster regrowth and an increased density of treated lashes [55]. For example, in a multicenter, randomized, double-blinded, parallel-group study in Japanese female chemotherapy patients, bimatoprost-treated subjects showed significantly greater increases in eyelash length, thickness, and darkness at 4 months compared to vehicle-treated subjects (p ≤ 0.04, N = 36) [56]. In another multicenter, randomized, double-blinded, parallel-group study in predominantly white female chemotherapy patients, improved global eyelash assessment was made in 37.5% of subjects receiving bimatoprost versus 18.2% of subjects receiving vehicle (p = 0.04) at month 4 and 46.9% versus 18.2% (p < 0.01) at month 6 [57]. Adverse effects include conjunctival hyperemia, punctate keratitis, and eye pruritus, and were largely localized to the treatment area, mild in severity, reversible upon treatment cessation, and predictable based on the known pharmacology of bimatoprost [57]. These studies and others provide statistically significant evidence that bimatoprost 0.03% application once daily to the upper eyelids promotes eyelash regrowth after CIA in multiple racial groups.
Additionally, latanoprost, another prostaglandin F2α analog used to treat increased pressure inside the eye, was also found to stimulate eyelash growth and pigmentation [58]. It was further shown to also stimulate hair growth in pattern baldness [59,60], though its ability to stimulate hair regrowth after CIA remains to be investigated.

3.3. Photobiomodulation Therapy (PBMT)

Photobiomodulation therapy (PBMT), or low-level laser therapy, is largely used in medicine and dentistry due to its ability to accelerate healing by increasing cell viability [61,62]. Recent studies have illustrated that PBMT stimulates mitochondrial cellular respiration via increasing ATP synthesis by photodissociation of the inhibitory nitric oxide from the heme and copper center of cytochrome C oxidase [61,62,63]. While PBMT has been more widely used for wound healing and reduction of pain and inflammation in musculoskeletal disorders, it has gained momentum in the 21st century as a potent treatment for hair loss by inducing HFs to exit telogen and enter an active anagen phase [61,62,64]. Because of its excellent safety profile, non-invasive nature, relatively low costs, availability as home devices, and ease to us, PBMT is a popular option for patients and is used with high compliance rates [64]. PBMT has been shown to be effective in treating male and female pattern hair loss and alopecia areata [61,62,64,65], but its effectiveness on CIA has not been extensively explored. It is hypothesized that PBMT for patients with CIA may inhibit apoptosis in the HFs by upregulating anti-apoptotic proteins such as those in the mitochondria [61,66]. While PBMT using red lights (630–655 nm) has shown successful results in promoting hair density and hair growth in patients as well as in animal models, recent studies have found positive effects of blue lights (453 nm) on ex vivo hair growth [66,67,68,69,70]. This could be due to the presence of another group of chromophores, called opsins, within the HFs. Opsins are responsive to blue-light signals, reduce apoptosis, and prolong anagen; however, future studies are needed to study the effect of blue light in vivo [61,66]. Thus, while PBMT has been shown to be successful in treating CIA in rat models [71] and other types of alopecia in human studies, further investigation is needed concerning human CIA. Results from three current clinical trials will help determine the safety and efficacy of PBMT in promoting hair regrowth after CIA (Table 2).

3.4. Platelet-Rich Plasma (PRP)

Platelet-rich plasma (PRP), also called autologous platelet concentrates or APCs, is a concentrate of platelet-rich plasma proteins derived from the whole blood, prepared by removing red blood cells and a portion of plasma by centrifugation, sometimes followed by sonication [72]. PRP is “activated” when growth factors and cytokines are released. Whereas host dermal collagen and endogenous thrombin are able to activate PRP, calcium gluconate, calcium chloride, or exogenous thrombin can be added before administration (“activated PRP” or AA-PRP) [73]. Activation helps to release many growth factors, including platelet-derived growth factor (PDGF) type a and b, transforming growth factor (TGF) type α and β, vascular endothelial growth factor (VEGF), epidermal growth factor (EGF), fibroblast growth factor (FGF), connective tissue growth factor (CTGF), and insulin-like growth factor-1 (IGF-1) [73]. Additionally, platelets can also release numerous anti-inflammatory cytokines such as interleukin-1 receptor antagonist (IL-1ra), soluble tumor necrosis factor (TNF) receptor (sTNF-R) I, interleukin (IL)-4, IL-10, IL-13, and interferon γ [73]. PRP has been used as an autologous injection for various medical conditions to promote healing and tissue regeneration/rejuvenation [73,74,75,76,77,78]. Additionally, PRP has also been used widely in alopecia treatment due to its ability to induce cell proliferation, supporting HF growth, and extending anagen [72,79,80,81,82]. Whereas PRP treatment has the advantages of minimal invasiveness, low cost, and being autologous, its effectiveness has not been vigorously validated, partly due to the autologous nature and partly due to the variations in preparation protocols [83]. A study in rats investigated the effects of clinically relevant concentration and preparation of PRP on preventing CIA by cytosine arabinoside (Ara-C), etoposide (VP-16), and Cytoxan (cyclophosphamide) chemotherapeutic agents, but did not show any protection against CIA or promotion of hair regrowth afterwards [84]. Therefore, the effectiveness of PRP needs to be vigorously validated before its use can be expanded in the clinic. Additionally, PRP is relatively contraindicated in patients with hematologic malignancies. A pilot study on the effectiveness of PRP for the treatment of endocrine therapy-induced alopecia and pCIA in breast cancer patients (NCT04459650) is expected to complete shortly.

3.5. Spironolactone

Spironolactone is a synthetic aldosterone receptor antagonist that has been used off-label for various dermatological conditions [85,86,87]. It inhibits circulating testosterone by occupying the androgen receptor and thereby displacing androgenic metabolites such as 5 alpha-dihydrotestosterone, thus limiting the nuclear translocation and subsequent transcriptional activation by androgen receptor. While its antiandrogenic properties prevent spironolactone from being used by males, it is suitable to treat conditions in females in which excess androgen production leads to unwanted and psychologically distressing manifestations, including acne, hidradenitis suppurativa, hirsutism, and female pattern hair loss [85,88,89,90,91,92]. A recent study also explored using spironolactone to treat pCIA [11]. In a group of 54 patients, most of whom had breast cancer and developed pCIA after taxane-based chemotherapy, 36 (67%) showed a moderate to significant improvement after treatment with oral spironolactone combined with topical minoxidil or topical minoxidil alone [11]. While this study did not assess the effects of spironolactone alone on pCIA, a separate study showed that spironolactone alone was effective in promoting hair growth and inhibiting alopecia progression, at a dose of 200 mg/day [89]. There had been concerns about spironolactone regarding the hypothetical risk of hormonal stimulation of endocrine receptor-positive tumors [93]; however, recent studies suggest that spironolactone might actually be linked with reduced cancer incidents [94,95,96,97,98,99]. Nonetheless, more extensive research is needed to confirm the effectiveness of spironolactone in treating CIA and assess potential adverse effects.

3.6. Other Agents

Besides the approaches discussed above, a variety of other topical and systemic agents have also been tested to prevent or treat CIA. Cyclosporine, an immunosuppressive calcineurin inhibitor that is known for inducing hypertrichosis, has been shown to not only induce active hair growth but also inhibits regression of HF in experimental models [100]. Murine studies have shown that with the use of etoposide, cytarabine, cyclophosphamide, and doxorubicin, both oral and topical cyclosporine can induce regrowth and slow hair loss, as well as prevent already damaged HFs from progressing into the terminal telogen phase [100]. Cyclosporine was also shown to prolong anagen in organ-cultured human HFs, suppressing the Wnt inhibitor SFRP1 in the dermal papilla [101]. In addition, topical calcineurin inhibitors such as tacrolimus and pimecrolimus have shown limited effectiveness in the treatment of alopecia areata [102] and frontal fibrosing alopecia [103,104]. Animal studies also showed that topical tacrolimus induces anagen and protects from CIA [105], although its effectiveness in patients remains to be assessed.
Furthermore, antioxidants are postulated to be effective against chemotherapy-induced toxicity due to their ability to eliminate free radicals and other reactive oxygen species created by chemotherapy [106,107]. In a rat model of CIA, topical application of edaravone ointment of 3% or higher led to decreased hair loss after cyclophosphamide chemotherapy [106]. Furthermore, when a novel antioxidant M30 [5-(N-methyl-N-propargylaminomethyl)-8-hydroxyquinoline]—a multi-target iron chelator—was given to mice injected with cyclophosphamide, elevated hair growth, as well as inhibited growth of cyclophosphamide-induced abnormal hair, were observed [108]. Moreover, 0.5–1% sodium zinc dihydrolipoylhistidinate (DHLHZn) applied topically to rat skin reduced the inflammatory cell infiltration around the HFs induced by cytosine arabinoside [109]. A more recent study in mouse models injected with cyclophosphamide showed that DHLHZn treatment preserved the level of insulin-like growth factor-1, an anagen promoter, and improved hair bulb diameter, leading to decreased alopecia [110].

3.7. Complementary and Alternative Treatments

Due to the varied efficacy and application regimen of current treatments for alopecia, many patients have turned to complementary and alternative medicine (CAM) for safe and natural alternatives. Many natural products have been claimed to increase hair density, with most studies conducted for androgenetic alopecia and alopecia areata. Natural products that have been suggested to be effective for alopecia treatment include amino acids (N-acetyl-L-cysteine, L-cystine, lysine), caffeine, capsaicin, curcumin, garlic gel, marine proteins (including extracellular matrix components from sharks and mollusks), melatonin, onion juice, procyanidin flavonoids, pumpkin seed oil, rosemary oil, saw palmetto, vitamin B6, vitamin B7 (biotin), vitamin D, vitamin E derivatives, zinc, and oral glucosides of peony with compound glycyrrhizin [111,112]. L-cystine combined with vitamin B6 have also been shown to effectively prevent doxorubicin-induced alopecia in mice [113]. In addition, hypnosis, essential oil aromatherapy, acupuncture, massage, electromagnetic stimulation, mindfulness psychotherapy, and homeopathy have also been proposed to improve psychologic and quality of life outcomes for alopecia patients [22,111,112]. Some of the studies on CAM effects lacked strong evidence, and more rigorous studies are needed to prove efficacy. Encouragingly, a topical lotion, CG428, made from botanical products was recently shown in a randomized double-blind controlled trial (NCT02605629) (Table 2) to promote hair density and thickness in breast cancer survivors with pCIA [114]. CG428 is a botanical product containing a patented blend of four botanical ingredients—citrus, cocoa, guarana, and onion [114]. After 6 months of application (twice daily), participants in the CG428 group showed a higher average hair density increase than the placebo group, and were more likely to report improvement of overall volume, hair density, and thickness, although these differences were not statistically significant [114]. It was speculated that CG428 may reduce apoptosis within the HFs through increasing the expression of anti-apoptotic Bcl-2, thereby prolonging the anagen phase and increasing the anagen:telogen HF ratio, allowing the new anagen hair to grow longer and to a thicker diameter [114]. Two other clinical trials on CG428 (NCT02919735 and NCT02986412) were recently completed, but the results are yet to be published (Table 2).

4. Supplementary Management

With hair loss often being seen as a sign of poor health or cancer treatment and its high incidence rate after chemotherapy [4], the anxiety and anticipation of chemotherapy causes tremendous psychological stress on patients, even influencing up to 14% of patients to consider rejecting chemotherapy and 8% of patients to completely reject chemotherapy [4,18,115]. Qualitative studies have also shown that many women experiencing breast cancer view CIA as being more distressing than losing a breast [116,117,118]. Thus, it is important to explore supplementary care and management through multidisciplinary approaches. Patients experiencing negative psychological concerns should be advised to seek psychological support through therapy or patient associations or support groups [21,119]. For aesthetic purposes, which play a large role in a patient’s QoL, other forms of supplementary management include the use of turbans, head scarfs, bandanas, and wigs [21,120]. Other camouflage techniques include cutting one’s hair shorter early on, keratin powder, and dermopigmentation [21,116,121,122]. Keratin powder uses natural keratin that attaches to a patients remaining hair or hair that is growing back via static electricity, making the hair appear fuller and denser temporarily [11,21]. Dermopigmentation is a pigment injection technique that deposits bioresorbable pigments into the epidermis and superficial dermis [21,121,122]. This is a temporary but long-lasting method that gives the illusion of increased hair density for 2 to 5 years and can be particularly useful to camouflage alopecia of the scalp or eyebrow [21]. When looking into the future, with the expected large increase in cancer survivors, it is important to find other methods of palliative care that can help increase a patient’s QoL while dealing with and recovering from CIA, taking into consideration the psychological impact of hair loss on these patients.

5. Conclusions

The current approaches to prevent and treat CIA are summarized in Figure 1. Some of the devices/agents are being studied in clinical trials (Table 2), and positive results may bring new options for cancer patients facing chemotherapy. Presently, scalp cooling is the most effective strategy to prevent CIA for patients with solid tumors undergoing certain chemotherapy regimen. Using FDA-cleared scalp cooling systems, scalp cooling has high patient compliance, and can reach a mean success rate of 50–70%. Self-administered cold caps, not cleared by the FDA, are also available from several manufacturers. However, scalp cooling is not suitable for patients with hematological tumors or certain peripheral neuropathies, or for patients prone to cold injury, and costs and access remain an issue. Even in countries with universal healthcare systems, the extra time needed for the patient to stay in the chemotherapy room is a drawback. For those patients who cannot use scalp cooling, application of topical agents such as calcitriol, KGF, and vasoconstrictors may offer good alternatives. Topical agents also have the advantage of being applied as often as daily instead of only during infusion time for scalp cooling.
For CIA treatment, minoxidil is by far the most effective agent to promote hair regrowth. Topical minoxidil has been found to shorten the alopecic period and promote hair regrowth after chemotherapy, while low-dose oral minoxidil has been shown to be an effective alternative for those who have difficulty using topical minoxidil. It is important to note that minoxidil should not be used during chemotherapy due to its vessel dilating effects but should be used after chemotherapy cessation. For eyelash regrowth, bimatoprost (0.03%) has shown efficacy in several studies, reporting increased eyelash length, thickness, and darkness in treated subjects compared to control. Additionally, several other treatment methods have been shown to promote hair regrowth in alopecia areata and androgenetic alopecia, and in animal studies against CIA. However, their effectiveness for treating CIA patients requires further investigation. Among them, PBMT stimulates mitochondrial cellular respiration and has been used for hair loss treatment by inducing HFs to enter an active anagen phase. PRP contains an abundance of growth factors that can induce cell proliferation and support HF growth, and therefore promote healing and tissue regeneration. CG428, spironolactone, cyclosporine, and antioxidants could potentially offer promising results in the treatment of CIA in the future. A combination of physical and chemical interventions may help to compensate for the shortcomings of either therapy alone. Additionally, given the importance of scalp and facial hair as a sign of good health, beauty, and youth, CIA has a significant negative impact on patients’ self-esteem, body image, sexuality, and overall QoL. Therefore, it is important to advise patients to seek psychological support and provide them with supplementary management methods such as wearing wigs, turbans, head scarfs, and using camouflage techniques.
Globally, new cancer cases are projected to reach 28.4 million in 2040 [123]. With prolonged life expectancy, earlier and more accurate diagnosis and more targeted treatment, the number of cancer survivors is expected to increase tremendously. In the U.S. alone, the number of cancer survivors is projected to grow from 18.1 million in 2022 to 26.0 million by 2040 [124]. Oncology teams who recognize the effects of CIA and work with dermatologists would better address CIA and provide better overall care to patients, and likely improve cancer treatment compliance and outcome.

Author Contributions

Conceptualization, T.C.W.; writing and editing, T.C.W., N.I.H., A.A., A.L.N. and M.M. All authors have read and agreed to the published version of the manuscript.

Funding

T.C.W. is an Endowed Frost Investigator, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine. This work is supported in part by AR079514 from NIH/NIAMS.

Acknowledgments

We would like to thank Ralf Paus and Xiangxi Michael Xu for helpful discussions.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. World Cancer Research Fund International. Worldwide Cancer Data. Available online: https://www.wcrf.org/cancer-trends/worldwide-cancer-data/ (accessed on 14 February 2023).
  2. Wilson, B.E.; Jacob, S.; Yap, M.L.; Ferlay, J.; Bray, F.; Barton, M.B. Estimates of global chemotherapy demands and corresponding physician workforce requirements for 2018 and 2040: A population-based study. Lancet Oncol. 2019, 20, 769–780. [Google Scholar] [CrossRef] [PubMed]
  3. Amarillo, D.; de Boni, D.; Cuello, M. Chemotherapy, Alopecia, and Scalp Cooling Systems. Actas Dermosifiliogr. 2022, 113, 278–283. [Google Scholar] [CrossRef]
  4. Freites-Martinez, A.; Shapiro, J.; Goldfarb, S.; Nangia, J.; Jimenez, J.J.; Paus, R.; Lacouture, M.E. Hair disorders in patients with cancer. J. Am. Acad. Dermatol. 2019, 80, 1179–1196. [Google Scholar] [CrossRef] [PubMed]
  5. Miller, K.D.; Nogueira, L.; Devasia, T.; Mariotto, A.B.; Yabroff, K.R.; Jemal, A.; Kramer, J.; Siegel, R.L. Cancer treatment and survivorship statistics, 2022. CA Cancer J. Clin. 2022, 72, 409–436. [Google Scholar] [CrossRef]
  6. Saraswat, N.; Chopra, A.; Sood, A.; Kamboj, P.; Kumar, S. A Descriptive Study to Analyze Chemotherapy-Induced Hair Loss and its Psychosocial Impact in Adults: Our Experience from a Tertiary Care Hospital. Indian Dermatol. Online J. 2019, 10, 426–430. [Google Scholar] [CrossRef] [PubMed]
  7. Trueb, R.M. Chemotherapy-induced alopecia. Semin. Cutan. Med. Surg. 2009, 28, 11–14. [Google Scholar] [CrossRef]
  8. Rossi, A.; Fortuna, M.C.; Caro, G.; Pranteda, G.; Garelli, V.; Pompili, U.; Carlesimo, M. Chemotherapy-induced alopecia management: Clinical experience and practical advice. J. Cosmet. Dermatol. 2017, 16, 537–541. [Google Scholar] [CrossRef]
  9. Rossi, A.; Caro, G.; Fortuna, M.C.; Pigliacelli, F.; D’Arino, A.; Carlesimo, M. Prevention and Treatment of Chemotherapy-Induced Alopecia. Dermatol. Pract. Concept. 2020, 10, e2020074. [Google Scholar] [CrossRef]
  10. Santos, T.S.; Hernandez Galvis, K.; Vano Galvan, S.; Saceda-Corralo, D. Post-chemotherapy alopecia: What the dermatologist needs to know. Int. J. Dermatol. 2021, 60, 1313–1317. [Google Scholar] [CrossRef]
  11. Freites-Martinez, A.; Chan, D.; Sibaud, V.; Shapiro, J.; Fabbrocini, G.; Tosti, A.; Cho, J.; Goldfarb, S.; Modi, S.; Gajria, D.; et al. Assessment of Quality of Life and Treatment Outcomes of Patients With Persistent Postchemotherapy Alopecia. JAMA Dermatol. 2019, 155, 724–728. [Google Scholar] [CrossRef]
  12. Kang, D.; Kim, I.R.; Choi, E.K.; Im, Y.H.; Park, Y.H.; Ahn, J.S.; Lee, J.E.; Nam, S.J.; Lee, H.K.; Park, J.H.; et al. Permanent Chemotherapy-Induced Alopecia in Patients with Breast Cancer: A 3-Year Prospective Cohort Study. Oncologist 2019, 24, 414–420. [Google Scholar] [CrossRef] [Green Version]
  13. Chan, J.; Adderley, H.; Alameddine, M.; Armstrong, A.; Arundell, D.; Fox, R.; Harries, M.; Lim, J.; Salih, Z.; Tetlow, C.; et al. Permanent hair loss associated with taxane chemotherapy use in breast cancer: A retrospective survey at two tertiary UK cancer centres. Eur. J. Cancer Care 2021, 30, e13395. [Google Scholar] [CrossRef]
  14. Bhoyrul, B.; Asfour, L.; Lutz, G.; Mitchell, L.; Jerjen, R.; Sinclair, R.D.; Holmes, S.; Chaudhry, I.H.; Harries, M.J. Clinicopathologic Characteristics and Response to Treatment of Persistent Chemotherapy-Induced Alopecia in Breast Cancer Survivors. JAMA Dermatol. 2021, 157, 1335–1342. [Google Scholar] [CrossRef] [PubMed]
  15. Piccini, I.; Brunken, L.; Cheret, J.; Ghatak, S.; Ramot, Y.; Alam, M.; Purba, T.S.; Hardman, J.; Erdmann, H.; Jimenez, F.; et al. Peroxisome proliferator-activated receptor-gamma signalling protects hair follicle stem cells from chemotherapy-induced apoptosis and epithelial-mesenchymal transition. Br. J. Dermatol. 2022, 186, 129–141. [Google Scholar] [CrossRef] [PubMed]
  16. Purba, T.S.; Ng’andu, K.; Brunken, L.; Smart, E.; Mitchell, E.; Hassan, N.; O’Brien, A.; Mellor, C.; Jackson, J.; Shahmalak, A.; et al. CDK4/6 inhibition mitigates stem cell damage in a novel model for taxane-induced alopecia. EMBO Mol. Med. 2019, 11, e11031. [Google Scholar] [CrossRef] [PubMed]
  17. Hadshiew, I.M.; Foitzik, K.; Arck, P.C.; Paus, R. Burden of hair loss: Stress and the underestimated psychosocial impact of telogen effluvium and androgenetic alopecia. J. Investig. Dermatol. 2004, 123, 455–457. [Google Scholar] [CrossRef] [Green Version]
  18. Paus, R.; Haslam, I.S.; Sharov, A.A.; Botchkarev, V.A. Pathobiology of chemotherapy-induced hair loss. Lancet Oncol. 2013, 14, e50–e59. [Google Scholar] [CrossRef]
  19. Gandhi, M.; Oishi, K.; Zubal, B.; Lacouture, M.E. Unanticipated toxicities from anticancer therapies: Survivors’ perspectives. Support. Care Cancer 2010, 18, 1461–1468. [Google Scholar] [CrossRef]
  20. Delgado Rodriguez, J.; Ramos-Garcia, V.; Infante-Ventura, D.; Suarez-Herrera, J.C.; Rueda-Dominguez, A.; Serrano-Aguilar, P.; Del Mar Trujillo-Martin, M. Ethical, legal, organizational and social issues related to the use of scalp cooling for the prevention of chemotherapy-induced alopecia: A systematic review. Health Expect. 2022, 26, 567–578. [Google Scholar] [CrossRef]
  21. Quesada, S.; Guichard, A.; Fiteni, F. Cancer-Related Alopecia: From Etiologies to Global Management. Cancers 2021, 13, 5556. [Google Scholar] [CrossRef] [PubMed]
  22. Haque, E.; Alabdaljabar, M.S.; Ruddy, K.J.; Haddad, T.C.; Thompson, C.A.; Lehman, J.S.; Hashmi, S.K. Management of chemotherapy-induced alopecia (CIA): A comprehensive review and future directions. Crit. Rev. Oncol. Hematol. 2020, 156, 103093. [Google Scholar] [CrossRef] [PubMed]
  23. Boyle, F.M.; Shaw, J.; Young, A.; van den Hurk, C.; Rugo, H.S.; Fogarty, G.B.; Lacouture, M.E. Management of Alopecia Due to Cancer Therapies; Olver, I., Ed.; Springer: Cham, Switzerland, 2018. [Google Scholar] [CrossRef]
  24. Olsen, E.A. Chemotherapy-Induced Alopecia: Overview and Methodology for Characterizing Hair Changes and Regrowth. In The MASCC Textbook of Cancer Supportive Care and Survivorship; Olver, I., Ed.; Springer: New York, NY, USA, 2011; pp. 381–386. [Google Scholar]
  25. Chen, L.; Xu, Y.; Ye, X. Low temperature mitigating the paclitaxel-induced damages in mouse cell and hair follicle model. Biochem. Biophys. Res. Commun. 2022, 603, 94–101. [Google Scholar] [CrossRef]
  26. Hillen, H.F.; Breed, W.P.; Botman, C.J. Scalp cooling by cold air for the prevention of chemotherapy-induced alopecia. Neth. J. Med. 1990, 37, 231–235. [Google Scholar] [PubMed]
  27. Wang, S.; Yang, T.; Shen, A.; Qiang, W.; Zhao, Z.; Zhang, F. The scalp cooling therapy for hair loss in breast cancer patients undergoing chemotherapy: A systematic review and meta-analysis. Support. Care Cancer 2021, 29, 6943–6956. [Google Scholar] [CrossRef] [PubMed]
  28. Marks, D.H.; Okhovat, J.P.; Hagigeorges, D.; Manatis-Lornell, A.J.; Isakoff, S.J.; Lacouture, M.E.; Senna, M.M. The effect of scalp cooling on CIA-related quality of life in breast cancer patients: A systematic review. Breast Cancer Res. Treat. 2019, 175, 267–276. [Google Scholar] [CrossRef] [PubMed]
  29. Kanat, O.; Ertas, H.; Caner, B. Platinum-induced neurotoxicity: A review of possible mechanisms. World J. Clin. Oncol. 2017, 8, 329–335. [Google Scholar] [CrossRef] [PubMed]
  30. Komen, M.M.; Smorenburg, C.H.; van den Hurk, C.J.; Nortier, J.W. Factors influencing the effectiveness of scalp cooling in the prevention of chemotherapy-induced alopecia. Oncologist 2013, 18, 885–891. [Google Scholar] [CrossRef] [Green Version]
  31. Saini, K.; Mysore, V. Role of vitamin D in hair loss: A short review. J. Cosmet. Dermatol. 2021, 20, 3407–3414. [Google Scholar] [CrossRef] [PubMed]
  32. Rashad, A.F.; Elgamal, E.; Fouda, I. Intralesional vitamin D3 in treatment of alopecia areata: A randomized controlled clinical trial. J. Cosmet. Dermatol. 2022, 21, 4617–4622. [Google Scholar] [CrossRef]
  33. Hidalgo, M.; Rinaldi, D.; Medina, G.; Griffin, T.; Turner, J.; Von Hoff, D.D. A phase I trial of topical topitriol (calcitriol, 1,25-dihydroxyvitamin D3) to prevent chemotherapy-induced alopecia. Anticancer Drugs 1999, 10, 393–395. [Google Scholar] [CrossRef] [PubMed]
  34. Lacouture, M.E.; Dion, H.; Ravipaty, S.; Jimenez, J.J.; Thapa, K.; Grund, E.M.; Benaim, E.; Tanna, N.; Luan, S.; DiTommaso, N.; et al. A phase I safety study of topical calcitriol (BPM31543) for the prevention of chemotherapy-induced alopecia. Breast Cancer Res. Treat. 2021, 186, 107–114. [Google Scholar] [CrossRef]
  35. Yen, T.T.; Thao, D.T.; Thuoc, T.L. An overview on keratinocyte growth factor: From the molecular properties to clinical applications. Protein Pept. Lett. 2014, 21, 306–317. [Google Scholar] [CrossRef]
  36. Kapoor, R.; Shome, D. Intradermal injections of a hair growth factor formulation for enhancement of human hair regrowth—Safety and efficacy evaluation in a first-in-man pilot clinical study. J. Cosmet. Laser Ther. 2018, 20, 369–379. [Google Scholar] [CrossRef]
  37. Danilenko, D.M.; Ring, B.D.; Yanagihara, D.; Benson, W.; Wiemann, B.; Starnes, C.O.; Pierce, G.F. Keratinocyte growth factor is an important endogenous mediator of hair follicle growth, development, and differentiation. Normalization of the nu/nu follicular differentiation defect and amelioration of chemotherapy-induced alopecia. Am. J. Pathol. 1995, 147, 145–154. [Google Scholar]
  38. Bodo, E.; Tobin, D.J.; Kamenisch, Y.; Biro, T.; Berneburg, M.; Funk, W.; Paus, R. Dissecting the impact of chemotherapy on the human hair follicle: A pragmatic in vitro assay for studying the pathogenesis and potential management of hair follicle dystrophy. Am. J. Pathol. 2007, 171, 1153–1167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Soref, C.M.; Fahl, W.E. A new strategy to prevent chemotherapy and radiotherapy-induced alopecia using topically applied vasoconstrictor. Int. J. Cancer 2015, 136, 195–203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Amaya, C.; Smith, E.R.; Xu, X.X. Low Intensity Ultrasound as an Antidote to Taxane/Paclitaxel-induced Cytotoxicity. J. Cancer 2022, 13, 2362–2373. [Google Scholar] [CrossRef] [PubMed]
  41. Amaya, C.; Luo, S.; Baigorri, J.; Baucells, R.; Smith, E.R.; Xu, X.X. Exposure to low intensity ultrasound removes paclitaxel cytotoxicity in breast and ovarian cancer cells. BMC Cancer 2021, 21, 981. [Google Scholar] [CrossRef]
  42. Stokes, G.S.; Oates, H.F.; MacCarthy, E.P. Antihypertensive therapy: New pharmacological approaches. Am. Heart J. 1980, 100, 741–752. [Google Scholar] [CrossRef]
  43. Rossi, A.; Anzalone, A.; Fortuna, M.C.; Caro, G.; Garelli, V.; Pranteda, G.; Carlesimo, M. Multi-therapies in androgenetic alopecia: Review and clinical experiences. Dermatol. Ther. 2016, 29, 424–432. [Google Scholar] [CrossRef] [Green Version]
  44. Rodriguez, R.; Machiavelli, M.; Leone, B.; Romero, A.; Cuevas, M.A.; Langhi, M.; Romero Acuna, L.; Romero Acuna, J.; Amato, S.; Barbieri, M.; et al. Minoxidil (Mx) as a prophylaxis of doxorubicin--induced alopecia. Ann. Oncol. 1994, 5, 769–770. [Google Scholar] [CrossRef]
  45. Stoehr, J.R.; Choi, J.N.; Colavincenzo, M.; Vanderweil, S. Off-Label Use of Topical Minoxidil in Alopecia: A Review. Am. J. Clin. Dermatol. 2019, 20, 237–250. [Google Scholar] [CrossRef] [PubMed]
  46. Randolph, M.; Tosti, A. Oral minoxidil treatment for hair loss: A review of efficacy and safety. J. Am. Acad. Dermatol. 2021, 84, 737–746. [Google Scholar] [CrossRef]
  47. Duvic, M.; Lemak, N.A.; Valero, V.; Hymes, S.R.; Farmer, K.L.; Hortobagyi, G.N.; Trancik, R.J.; Bandstra, B.A.; Compton, L.D. A randomized trial of minoxidil in chemotherapy-induced alopecia. J. Am. Acad. Dermatol. 1996, 35, 74–78. [Google Scholar] [CrossRef] [PubMed]
  48. Lyakhovitsky, A.; Segal, O.; Maly, A.; Zlotogorski, A.; Barzilai, A. Permanent chemotherapy-induced alopecia after hematopoietic stem cell transplantation treated with low-dose oral minoxidil. JAAD Case Rep. 2022, 22, 64–67. [Google Scholar] [CrossRef] [PubMed]
  49. Chen, Y.F.; Chen, L.H.; Yeh, Y.M.; Wu, P.Y.; Chen, Y.F.; Chang, L.Y.; Chang, J.Y.; Shen, M.R. Minoxidil is a potential neuroprotective drug for paclitaxel-induced peripheral neuropathy. Sci. Rep. 2017, 7, 45366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Zhou, L.; Zhan, W.; Wei, X. Clinical pharmacology and pharmacogenetics of prostaglandin analogues in glaucoma. Front. Pharmacol. 2022, 13, 1015338. [Google Scholar] [CrossRef]
  51. Mohan, N.; Chakrabarti, A.; Nazm, N.; Mehta, R.; Edward, D.P. Newer advances in medical management of glaucoma. Indian J. Ophthalmol. 2022, 70, 1920–1930. [Google Scholar] [CrossRef]
  52. Herane, M.I.; Urbina, F. Acquired trichomegaly of the eyelashes and hypertrichosis induced by bimatoprost. J. Eur. Acad. Dermatol. Venereol. 2004, 18, 644–645. [Google Scholar] [CrossRef] [PubMed]
  53. Hart, J.; Shafranov, G. Hypertrichosis of vellus hairs of the malar region after unilateral treatment with bimatoprost. Am. J. Ophthalmol. 2004, 137, 756–757. [Google Scholar] [CrossRef]
  54. Law, S.K. Bimatoprost in the treatment of eyelash hypotrichosis. Clin. Ophthalmol. 2010, 4, 349–358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Barron-Hernandez, Y.L.; Tosti, A. Bimatoprost for the treatment of eyelash, eyebrow and scalp alopecia. Expert Opin. Investig. Drugs 2017, 26, 515–522. [Google Scholar] [CrossRef] [PubMed]
  56. Harii, K.; Arase, S.; Tsuboi, R.; Weng, E.; Daniels, S.; VanDenburgh, A. Bimatoprost for eyelash growth in Japanese subjects: Two multicenter controlled studies. Aesthetic Plast. Surg. 2014, 38, 451–460. [Google Scholar] [CrossRef] [Green Version]
  57. Glaser, D.A.; Hossain, P.; Perkins, W.; Griffiths, T.; Ahluwalia, G.; Weng, E.; Beddingfield, F.C. Long-term safety and efficacy of bimatoprost solution 0.03% application to the eyelid margin for the treatment of idiopathic and chemotherapy-induced eyelash hypotrichosis: A randomized controlled trial. Br. J. Dermatol. 2015, 172, 1384–1394. [Google Scholar] [CrossRef] [Green Version]
  58. Coondoo, A.; Sengupta, S. Serendipity and its role in dermatology. Indian J. Dermatol. 2015, 60, 130–135. [Google Scholar] [CrossRef] [PubMed]
  59. Blume-Peytavi, U.; Lonnfors, S.; Hillmann, K.; Garcia Bartels, N. A randomized double-blind placebo-controlled pilot study to assess the efficacy of a 24-week topical treatment by latanoprost 0.1% on hair growth and pigmentation in healthy volunteers with androgenetic alopecia. J. Am. Acad. Dermatol. 2012, 66, 794–800. [Google Scholar] [CrossRef]
  60. Levy, L.L.; Emer, J.J. Female pattern alopecia: Current perspectives. Int. J. Womens Health 2013, 5, 541–556. [Google Scholar] [CrossRef] [Green Version]
  61. Hamblin, M.R. Photobiomodulation for the management of alopecia: Mechanisms of action, patient selection and perspectives. Clin. Cosmet. Investig. Dermatol. 2019, 12, 669–678. [Google Scholar] [CrossRef] [Green Version]
  62. Dompe, C.; Moncrieff, L.; Matys, J.; Grzech-Lesniak, K.; Kocherova, I.; Bryja, A.; Bruska, M.; Dominiak, M.; Mozdziak, P.; Skiba, T.H.I.; et al. Photobiomodulation-Underlying Mechanism and Clinical Applications. J. Clin. Med. 2020, 9, 1724. [Google Scholar] [CrossRef] [PubMed]
  63. Lane, N. Cell biology: Power games. Nature 2006, 443, 901–903. [Google Scholar] [CrossRef]
  64. da Silveira, S.P.; Moita, S.R.U.; da Silva, S.V.; Rodrigues, M.; da Silva, D.F.T.; Pavani, C. The role of photobiomodulation when associated with microneedling in female pattern hair loss: A randomized, double blind, parallel group, three arm, clinical study protocol. Medicine (Baltimore) 2019, 98, e14938. [Google Scholar] [CrossRef] [PubMed]
  65. Jimenez, J.J.; Wikramanayake, T.C.; Bergfeld, W.; Hordinsky, M.; Hickman, J.G.; Hamblin, M.R.; Schachner, L.A. Efficacy and safety of a low-level laser device in the treatment of male and female pattern hair loss: A multicenter, randomized, sham device-controlled, double-blind study. Am. J. Clin. Dermatol. 2014, 15, 115–127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Buscone, S.; Mardaryev, A.N.; Raafs, B.; Bikker, J.W.; Sticht, C.; Gretz, N.; Farjo, N.; Uzunbajakava, N.E.; Botchkareva, N.V. A new path in defining light parameters for hair growth: Discovery and modulation of photoreceptors in human hair follicle. Lasers Surg. Med. 2017, 49, 705–718. [Google Scholar] [CrossRef] [Green Version]
  67. Lanzafame, R.J.; Blanche, R.R.; Bodian, A.B.; Chiacchierini, R.P.; Fernandez-Obregon, A.; Kazmirek, E.R. The growth of human scalp hair mediated by visible red light laser and LED sources in males. Lasers Surg. Med. 2013, 45, 487–495. [Google Scholar] [CrossRef] [PubMed]
  68. Lanzafame, R.J.; Blanche, R.R.; Chiacchierini, R.P.; Kazmirek, E.R.; Sklar, J.A. The growth of human scalp hair in females using visible red light laser and LED sources. Lasers Surg. Med. 2014, 46, 601–607. [Google Scholar] [CrossRef]
  69. Avci, P.; Gupta, G.K.; Clark, J.; Wikonkal, N.; Hamblin, M.R. Low-level laser (light) therapy (LLLT) for treatment of hair loss. Lasers Surg. Med. 2014, 46, 144–151. [Google Scholar] [CrossRef] [Green Version]
  70. Luck, M.; Mathes, T.; Bruun, S.; Fudim, R.; Hagedorn, R.; Tran Nguyen, T.M.; Kateriya, S.; Kennis, J.T.; Hildebrandt, P.; Hegemann, P. A photochromic histidine kinase rhodopsin (HKR1) that is bimodally switched by ultraviolet and blue light. J. Biol. Chem. 2012, 287, 40083–40090. [Google Scholar] [CrossRef] [Green Version]
  71. Wikramanayake, T.C.; Villasante, A.C.; Mauro, L.M.; Nouri, K.; Schachner, L.A.; Perez, C.I.; Jimenez, J.J. Low-level laser treatment accelerated hair regrowth in a rat model of chemotherapy-induced alopecia (CIA). Lasers Med. Sci. 2013, 28, 701–706. [Google Scholar] [CrossRef]
  72. Alves, R.; Grimalt, R. A Review of Platelet-Rich Plasma: History, Biology, Mechanism of Action, and Classification. Ski. Appendage Disord. 2018, 4, 18–24. [Google Scholar] [CrossRef]
  73. Mercuri, S.R.; Paolino, G.; Di Nicola, M.R.; Vollono, L. Investigating the Safety and Efficacy of Platelet-Rich Plasma (PRP) Treatment for Female Androgenetic Alopecia: Review of the Literature. Medicina (Kaunas) 2021, 57, 311. [Google Scholar] [CrossRef]
  74. Verma, R.; Kumar, S.; Garg, P.; Verma, Y.K. Platelet-rich plasma: A comparative and economical therapy for wound healing and tissue regeneration. Cell Tissue Bank. 2022, 1–22. [Google Scholar] [CrossRef]
  75. Ebrahimi, Z.; Alimohamadi, Y.; Janani, M.; Hejazi, P.; Kamali, M.; Goodarzi, A. Platelet-rich plasma in the treatment of scars, to suggest or not to suggest? A systematic review and meta-analysis. J. Tissue Eng. Regen. Med. 2022, 16, 875–899. [Google Scholar] [CrossRef] [PubMed]
  76. O’Sullivan, L.; Ni Riordain, R. Autologous platelet concentrates in oral surgery: Protocols, properties, and clinical applications. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2022, 133, 156–164. [Google Scholar] [CrossRef] [PubMed]
  77. Cecerska-Heryc, E.; Goszka, M.; Serwin, N.; Roszak, M.; Grygorcewicz, B.; Heryc, R.; Dolegowska, B. Applications of the regenerative capacity of platelets in modern medicine. Cytokine Growth Factor Rev. 2022, 64, 84–94. [Google Scholar] [CrossRef] [PubMed]
  78. Mercuri, S.R.; Di Nicola, M.R.; Brianti, P.; Bianchi, V.G.; Paolino, G. Pilot Study on the Use of the “Monocyte-Rich” Platelet-Rich Plasma in Combination with 1927 nm Fractional and 308 nm Excimer Lasers for the Treatment of Vitiligo. Medicina (Kaunas) 2021, 57, 904. [Google Scholar] [CrossRef] [PubMed]
  79. Ayatollahi, A.; Hosseini, H.; Gholami, J.; Mirminachi, B.; Firooz, F.; Firooz, A. Platelet rich plasma for treatment of non-scarring hair loss: Systematic review of literature. J. Dermatolog. Treat 2017, 28, 574–581. [Google Scholar] [CrossRef] [PubMed]
  80. Sikora, M.; Rudnicka, L. Chemotherapy-induced alopecia—The urgent need for treatment options. J. Eur. Acad. Dermatol. Venereol. 2019, 33, e69–e70. [Google Scholar] [CrossRef]
  81. Paichitrojjana, A.; Paichitrojjana, A. Platelet Rich Plasma and Its Use in Hair Regrowth: A Review. Drug Des. Dev. Ther. 2022, 16, 635–645. [Google Scholar] [CrossRef]
  82. Shimizu, Y.; Ntege, E.H.; Sunami, H.; Inoue, Y. Regenerative medicine strategies for hair growth and regeneration: A narrative review of literature. Regen. Ther. 2022, 21, 527–539. [Google Scholar] [CrossRef]
  83. Cruciani, M.; Masiello, F.; Pati, I.; Marano, G.; Pupella, S.; De Angelis, V. Platelet-rich plasma for the treatment of alopecia: A systematic review and meta-analysis. Blood Transfus. 2021, 21, 24–36. [Google Scholar] [CrossRef]
  84. Stamatiou, C.A.; Lens, A.; Perez, C.I.; Daunert, S.; Jimenez, J.J. The Role of Platelet-Rich Plasma in the Prevention of Chemotherapy-Induced Alopecia. Ski. Appendage Disord. 2020, 6, 58–60. [Google Scholar] [CrossRef]
  85. Searle, T.N.; Al-Niaimi, F.; Ali, F.R. Spironolactone in dermatology: Uses in acne and beyond. Clin. Exp. Dermatol. 2020, 45, 986–993. [Google Scholar] [CrossRef] [PubMed]
  86. Aguilar Medina, D.A.; Cazarin, J.; Magana, M. Spironolactone in dermatology. Dermatol. Ther. 2022, 35, e15321. [Google Scholar] [CrossRef]
  87. Butt, A.K.; Patel, J.; Shirwany, H.; Mirza, Q.; Hoover, J.; Khouzam, R.N. Beneficial Extracardiac Effects of Cardiovascular Medications. Curr. Cardiol. Rev. 2022, 18, e151021197270. [Google Scholar] [CrossRef] [PubMed]
  88. Burns, L.J.; De Souza, B.; Flynn, E.; Hagigeorges, D.; Senna, M.M. Spironolactone for treatment of female pattern hair loss. J. Am. Acad. Dermatol. 2020, 83, 276–278. [Google Scholar] [CrossRef] [PubMed]
  89. Fabbrocini, G.; Cantelli, M.; Masara, A.; Annunziata, M.C.; Marasca, C.; Cacciapuoti, S. Female pattern hair loss: A clinical, pathophysiologic, and therapeutic review. Int. J. Womens Dermatol. 2018, 4, 203–211. [Google Scholar] [CrossRef] [PubMed]
  90. James, J.F.; Jamerson, T.A.; Aguh, C. Efficacy and safety profile of oral spironolactone use for androgenic alopecia: A systematic review. J. Am. Acad. Dermatol. 2022, 86, 425–429. [Google Scholar] [CrossRef] [PubMed]
  91. Sinclair, R.D. Female pattern hair loss: A pilot study investigating combination therapy with low-dose oral minoxidil and spironolactone. Int. J. Dermatol. 2018, 57, 104–109. [Google Scholar] [CrossRef]
  92. Vargas-Mora, P.; Morgado-Carrasco, D. Spironolactone in Dermatology: Uses in Acne, Hidradenitis Suppurativa, Female Pattern Baldness, and Hirsutism. Actas Dermosifiliogr. (Engl. Ed.) 2020, 111, 639–649. [Google Scholar] [CrossRef]
  93. Rozner, R.N.; Freites-Martinez, A.; Shapiro, J.; Geer, E.B.; Goldfarb, S.; Lacouture, M.E. Safety of 5alpha-reductase inhibitors and spironolactone in breast cancer patients receiving endocrine therapies. Breast Cancer Res. Treat 2019, 174, 15–26. [Google Scholar] [CrossRef]
  94. Funder, J.W. Spironolactone in cardiovascular disease: An expanding universe? F1000Research 2017, 6, 1738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Bommareddy, K.; Hamade, H.; Lopez-Olivo, M.A.; Wehner, M.; Tosh, T.; Barbieri, J.S. Association of Spironolactone Use With Risk of Cancer: A Systematic Review and Meta-analysis. JAMA Dermatol. 2022, 158, 275–282. [Google Scholar] [CrossRef] [PubMed]
  96. Mackenzie, I.S.; Morant, S.V.; Wei, L.; Thompson, A.M.; MacDonald, T.M. Spironolactone use and risk of incident cancers: A retrospective, matched cohort study. Br. J. Clin. Pharmacol. 2017, 83, 653–663. [Google Scholar] [CrossRef] [Green Version]
  97. Hiebert, B.M.; Janzen, B.W.; Sanjanwala, R.M.; Ong, A.D.; Feldman, R.D.; Kim, J.O. Impact of spironolactone exposure on prostate cancer incidence amongst men with heart failure: A Pharmacoepidemiological study. Br. J. Clin. Pharmacol. 2021, 87, 1801–1813. [Google Scholar] [CrossRef] [PubMed]
  98. Gold, A.; Eini, L.; Nissim-Rafinia, M.; Viner, R.; Ezer, S.; Erez, K.; Aqaqe, N.; Hanania, R.; Milyavsky, M.; Meshorer, E.; et al. Spironolactone inhibits the growth of cancer stem cells by impairing DNA damage response. Oncogene 2019, 38, 3103–3118. [Google Scholar] [CrossRef]
  99. Beckmann, K.; Garmo, H.; Lindahl, B.; Holmberg, L.; Stattin, P.; Adolfsson, J.; Cruickshank, J.K.; Van Hemelrijck, M. Spironolactone use is associated with lower prostate cancer risk: A population-wide case-control study. Prostate Cancer Prostatic Dis. 2020, 23, 527–533. [Google Scholar] [CrossRef] [PubMed]
  100. Dmytriw, A.A.; Morzycki, W.; Green, P.J. Prevention of alopecia in medical and interventional chemotherapy patients. J. Cutan. Med. Surg. 2015, 19, 11–16. [Google Scholar] [CrossRef] [PubMed]
  101. Hawkshaw, N.J.; Hardman, J.A.; Haslam, I.S.; Shahmalak, A.; Gilhar, A.; Lim, X.; Paus, R. Identifying novel strategies for treating human hair loss disorders: Cyclosporine A suppresses the Wnt inhibitor, SFRP1, in the dermal papilla of human scalp hair follicles. PLoS Biol. 2018, 16, e2003705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Barton, V.R.; Toussi, A.; Awasthi, S.; Kiuru, M. Treatment of pediatric alopecia areata: A systematic review. J. Am. Acad. Dermatol. 2022, 86, 1318–1334. [Google Scholar] [CrossRef] [PubMed]
  103. Heppt, M.V.; Letule, V.; Laniauskaite, I.; Reinholz, M.; Tietze, J.K.; Wolff, H.; Ruzicka, T.; Sattler, E.C. Frontal Fibrosing Alopecia: A Retrospective Analysis of 72 Patients from a German Academic Center. Facial Plast. Surg. 2018, 34, 88–94. [Google Scholar] [CrossRef]
  104. Gamret, A.C.; Potluri, V.S.; Krishnamurthy, K.; Fertig, R.M. Frontal fibrosing alopecia: Efficacy of treatment modalities. Int. J. Womens Health 2019, 11, 273–285. [Google Scholar] [CrossRef] [Green Version]
  105. De Tran, Q.H.; Guay, E.; Chartier, S.; Tousignant, J. Tacrolimus in dermatology. J. Cutan. Med. Surg. 2001, 5, 329–335. [Google Scholar] [CrossRef] [PubMed]
  106. Yoneda, K.; Fujii, M.; Imaoka, A.; Kobayashi, R.; Hayashi, R.; Yoshida, Y.; Kohno, T.; Tsuji, T. Preventive effect of edaravone ointment on cyclophosphamide-chemotherapy induced alopecia. Support. Care Cancer 2021, 29, 6127–6134. [Google Scholar] [CrossRef]
  107. Singh, K.; Bhori, M.; Kasu, Y.A.; Bhat, G.; Marar, T. Antioxidants as precision weapons in war against cancer chemotherapy induced toxicity—Exploring the armoury of obscurity. Saudi Pharm. J. 2018, 26, 177–190. [Google Scholar] [CrossRef]
  108. Lim, Y.C.; Kim, H.; Lim, S.M.; Kim, J.S. Genetic analysis of a novel antioxidant multi-target iron chelator, M30 protecting against chemotherapy-induced alopecia in mice. BMC Cancer 2019, 19, 149. [Google Scholar] [CrossRef]
  109. Hagiwara, S.; Uchida, T.; Koga, H.; Inomata, M.; Yoshizumi, F.; Moriyama, M.; Kitano, S.; Noguchi, T. The alpha-lipoic acid derivative sodium zinc dihydrolipoylhistidinate reduces chemotherapy-induced alopecia in a rat model: A pilot study. Surg. Today 2011, 41, 693–697. [Google Scholar] [CrossRef]
  110. Aiba, T.; Kono, Y.; Etoh, T.; Kawano, Y.; Oshima, Y.; Inomata, M. Efficacy of cooling therapy and alpha-lipoic acid derivative against chemotherapy-induced alopecia in an animal model. Cancer Sci. 2023, 114, 1007–1014. [Google Scholar] [CrossRef]
  111. Tkachenko, E.; Okhovat, J.P.; Manjaly, P.; Huang, K.P.; Senna, M.M.; Mostaghimi, A. Complementary and alternative medicine for alopecia areata: A systematic review. J. Am. Acad. Dermatol. 2023, 88, 131–143. [Google Scholar] [CrossRef]
  112. Hosking, A.M.; Juhasz, M.; Atanaskova Mesinkovska, N. Complementary and Alternative Treatments for Alopecia: A Comprehensive Review. Ski. Appendage Disord 2019, 5, 72–89. [Google Scholar] [CrossRef] [PubMed]
  113. D’Agostini, F.; Fiallo, P.; Ghio, M.; De Flora, S. Chemoprevention of doxorubicin-induced alopecia in mice by dietary administration of L-cystine and vitamin B6. Arch. Dermatol. Res. 2013, 305, 25–34. [Google Scholar] [CrossRef] [PubMed]
  114. Kang, D.; Kim, I.R.; Park, Y.H.; Im, Y.H.; Zhao, D.; Guallar, E.; Ahn, J.S.; Cho, J. Impact of a topical lotion, CG428, on permanent chemotherapy-induced alopecia in breast cancer survivors: A pilot randomized double-blind controlled clinical trial (VOLUME RCT). Support. Care Cancer 2020, 28, 1829–1837. [Google Scholar] [CrossRef] [PubMed]
  115. Frith, H.; Harcourt, D.; Fussell, A. Anticipating an altered appearance: Women undergoing chemotherapy treatment for breast cancer. Eur. J. Oncol. Nurs. 2007, 11, 385–391. [Google Scholar] [CrossRef]
  116. Boland, V.; Brady, A.M.; Drury, A. The physical, psychological and social experiences of alopecia among women receiving chemotherapy: An integrative literature review. Eur. J. Oncol. Nurs. 2020, 49, 101840. [Google Scholar] [CrossRef]
  117. Browall, M.; Gaston-Johansson, F.; Danielson, E. Postmenopausal women with breast cancer: Their experiences of the chemotherapy treatment period. Cancer Nurs. 2006, 29, 34–42. [Google Scholar] [CrossRef]
  118. Freedman, T.G. Social and cultural dimensions of hair loss in women treated for breast cancer. Cancer Nurs. 1994, 17, 334–341. [Google Scholar] [CrossRef]
  119. Carelle, N.; Piotto, E.; Bellanger, A.; Germanaud, J.; Thuillier, A.; Khayat, D. Changing patient perceptions of the side effects of cancer chemotherapy. Cancer 2002, 95, 155–163. [Google Scholar] [CrossRef] [PubMed]
  120. Freites-Martinez, A.; Shapiro, J.; van den Hurk, C.; Goldfarb, S.; Jimenez, J.J.; Rossi, A.M.; Paus, R.; Lacouture, M.E. Hair disorders in cancer survivors. J. Am. Acad. Dermatol. 2019, 80, 1199–1213. [Google Scholar] [CrossRef]
  121. Masser, M.R.; Di Meo, L.; Hobby, J.A. Tattooing in reconstruction of the nipple and areola: A new method. Plast. Reconstr. Surg. 1989, 84, 677–681. [Google Scholar] [CrossRef]
  122. Uhlmann, N.R.; Martins, M.M.; Piato, S. 3D areola dermopigmentation (nipple-areola complex). Breast J. 2019, 25, 1214–1221. [Google Scholar] [CrossRef] [PubMed]
  123. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  124. American Cancer Society. Cancer Treatment & Survivorship Facts & Figures 2022–2024; American Cancer Society: Atlanta, GA, USA, 2022. [Google Scholar]
Figure 1. Current approaches to prevent and treat CIA.
Figure 1. Current approaches to prevent and treat CIA.
Curroncol 30 00275 g001
Table 1. Comparison of the different scalp cooling devices used by patients with solid tumors undergoing chemotherapy.
Table 1. Comparison of the different scalp cooling devices used by patients with solid tumors undergoing chemotherapy.
DeviceDate of
FDA Clearance
Device DescriptionTemperature Control
by Computer
AdvantagesAdverse
Effects
OperatorManufacturerCosts
DigniCap C3 Scalp Cooling
System
2015 (for breast cancer) *mobile cooling unit that uses circulating cold coolantX• The wrap is
fitted at the
beginning of treatment and
remains on until completion
• Starting from room temperature: comfortable
feeling of
coldness, headache, scalp pain and/or light-headedness, dizziness
health-care
providers
administered
Dignitana~USD 1500–2000
DigniCap Delta Scalp Cooling
System
06/27/19Digitana~USD 1500–2000
Paxman Scalp Cooling System04/19/17, later
expanded to solid tumors
Paxman Coolers Limitedcapped at USD 2200
Amma Cooler Heads cold cap12/08/21Cooler Heads~USD 2000 rental costs
Penguin Cold Capsnot yet **3-cap system: Crylon Gel in the caps no• Portable
• Caps chill fast
self-administeredPenguin Cold
Caps
USD 419/month rental costs
Chemo Cold
Caps
not yet6-cap system: caps filled with coolant gelArctic Cold
Caps LLC
USD 379/month rental costs
Arctic Cold
Caps
not yet8-cap system: caps filled with glycerin-based hydr0-gel that refreezes in 2 hArctic Cold
Caps LLC
USD 379/month rental costs
* 07/03/17 FDA-cleared for expanded use in cancer patients with solid tumors. ** All materials used in the manufacture of Penguin Cold Caps are approved in Canada and by the FDA in the USA.
Table 2. Current clinical trials for the prevention and treatment of chemotherapy-induced alopecia (clinicaltrials.gov).
Table 2. Current clinical trials for the prevention and treatment of chemotherapy-induced alopecia (clinicaltrials.gov).
CategoryNCT
Number
InterventionsCompletedRecruitingNot Yet RecruitingDeviceRCT, Parallel
Assignment
Single Group
Assignment
Open LabelGenderTarget Sample Size/EnrollmentPreventionTreatment
Scalp coolingNCT01831024Dignicap System* XNon-R CT XFemale110X
NCT03712696Device: DIGNICAP™X X XXFemale139X
NCT04630080Scalp cooling X X XXFemale100X
NCT05213936Scalp cooling with hairstyle using conditioner and water emulsion X XNon-R CT Single blind (Investigator)All30X
NCT03248193Concomitant limb
cryocompression and scalp cooling
* XNon-R CT XAll50X
NCT04986579Paxman Scalp Cooling System X XNon-R CT XAll120X
NCT01008774Paxman Cooling
Machine; Cold Caps
X XNon-R CT XAll239X
NCT04180579PAXMAN Scalp Cooler **X XXFemale34X
NCT04764357Paxman Scalp Cooling System X X XXAll40X
NCT04117815Paxman Scalp Cooling SystemX X*** Female128X
NCT04626895Paxman Scalp Cooling DeviceX XX XFemale15X
NCT05533320Paxman Scalp Cooling System XX XXFemale30X
NCT04678544Paxman Scalp Cooling System 2X XX Single blind (Outcomes Assessor)Female170X
NCT04168242Scalp cooling Paxman
Orbis II system
**XX XFemale80X
NCT01986140PAXMAN Orbis Scalp Cooler Treatment with Orbis scalp cooling cap **XX Single blind (Care Provider)Female236X
NCT03289364Penguin Cold Caps* X XXAll9
NCT05484973AMMA Portable Scalp Cooling System XX XXFemale125X
NCT05365243AMMA Portable Scalp Cooling System XX XXFemale12X
NCT03711877Scalp cooling system; chemical cold cap X XX XFemale256X
PBMTNCT05177289Theradome® LH80 pro combined with scalp cooling X XX Single blind (Outcomes Assessor)Female72XX
NCT04036994Photobiomodulation therapy X XX Single blind (Participant)Female30 X
NCT05397457Low-level light therapy X XX XFemale88 X
TopicalNCT02919735Topical CG 428 herbal medicinal solutionX X Double blind (Participant, Investigator)Female40XX
NCT02986412Topical CG 428 herbal
lotion
X XXFemale19 X
NCT02605629Topical CG 428 herbal
lotion
X X Double blind (Participant, Investigator)Female32 X
NCT04554732Topical Keratinocyte growth factorX XXFemale28X
NCT01588522Topical compound 31, 543 CalcitriolX XXAll30X
OralNCT03831334Drug: oral minoxidil X XXAll25 X
InjectionNCT04459650Platelet Rich Plasma ** XXAll30 X
-NCT02530177Clinical Assessment of
alopecia and pCIA, skin
aging and nail changes: Observational
** **** Female546--
*: Unknown status; **: Active, not recruiting; ***: Observational Model: Cohort; ****: Observational Model: Case-Control. RCT: Randomized controlled trial; Non-R CT: Non-randomized controlled trial. Note: NCT00999557 to evaluate the efficacy of topical Bimatoprost in promoting eyebrow and eyelashes has been withdrawn. NCT02797223, an interview-based quality of life study supposed to be completed by December 2016, was excluded.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wikramanayake, T.C.; Haberland, N.I.; Akhundlu, A.; Laboy Nieves, A.; Miteva, M. Prevention and Treatment of Chemotherapy-Induced Alopecia: What Is Available and What Is Coming? Curr. Oncol. 2023, 30, 3609-3626. https://doi.org/10.3390/curroncol30040275

AMA Style

Wikramanayake TC, Haberland NI, Akhundlu A, Laboy Nieves A, Miteva M. Prevention and Treatment of Chemotherapy-Induced Alopecia: What Is Available and What Is Coming? Current Oncology. 2023; 30(4):3609-3626. https://doi.org/10.3390/curroncol30040275

Chicago/Turabian Style

Wikramanayake, Tongyu C., Nicole I. Haberland, Aysun Akhundlu, Andrea Laboy Nieves, and Mariya Miteva. 2023. "Prevention and Treatment of Chemotherapy-Induced Alopecia: What Is Available and What Is Coming?" Current Oncology 30, no. 4: 3609-3626. https://doi.org/10.3390/curroncol30040275

Article Metrics

Back to TopTop