ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Alopecia related to systemic cancer therapy

Alopecia related to systemic cancer therapy
Literature review current through: Jan 2024.
This topic last updated: Aug 03, 2023.

INTRODUCTION — Alopecia is a transient and usually (although not always) reversible consequence of systemic cancer therapy that can be psychologically and socially devastating [1]. Alopecia is often cited as one of the most negative effects on quality of life in patients with cancer [2]. For some patients, the emotional trauma may be so severe as to lead to choosing suboptimal treatment or refusing or delaying treatment that might otherwise be beneficial [3-10]. Recovery generally requires a period of several months to a year, amplifying the impact of the disease and its treatment.

A general overview of the anatomy and physiology of hair growth, the effects of systemic cancer therapies on the hair follicle, and possible means for preventing or minimizing alopecia are discussed here.

ANATOMY AND PHYSIOLOGY — The hair shaft is a layered structure that consists of three major components. The medulla, the innermost layer, is surrounded by the cortex and cuticle. The hair shaft is the product of the hair follicle, which is composed of three main parts when viewed in longitudinal section (figure 1) [11]:

The lower portion, which extends from the base of the hair follicle to the insertion of the arrector pili muscle. This lower portion, in turn, is comprised of several major components:

The hair bulb, which contains the dermal papilla and hair matrix. The dermal papilla controls the number of matrix cells, which determines hair fiber size [12]. Melanocytes, which are responsible for hair color, are present among the matrix cells of the hair bulb.

The hair shaft itself, consisting of medulla, cortex, and hair cuticle (inside to outside).

The inner root sheath, which consists of the inner root sheath cuticle, Huxley layer, and Henle layer (inside to outside). The inner root sheath is rigid, with its shape determining whether hair is curly or straight.

The outer root sheath.

Damage to the lower portion of the hair follicle, as occurs in autoimmune alopecia areata, can result in a nonscarring alopecia. Immune-mediated alopecia areata can also be induced by immune checkpoint inhibitors that target cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and programmed cell death 1 (PD-1)/programmed cell death 1 ligand (PD-L1) [13]. (See "Evaluation and diagnosis of hair loss", section on 'Nonscarring alopecia' and "Toxicities associated with immune checkpoint inhibitors" and "Alopecia areata: Clinical manifestations and diagnosis".)

The middle portion (isthmus), which extends from the insertion of the arrector pili muscle to the entrance of the sebaceous duct. This portion contains the "bulge" of the hair follicle, where the epithelial stem cells reside [14]. Damage to the bulge of the hair follicle results in irreversible scarring alopecia, as is seen clinically in disorders such as discoid lupus and lichen planopilaris.

The upper portion (infundibulum), which extends from the entrance of the sebaceous duct to the follicular orifice.

Once formed, hair follicles undergo lifelong cycling characterized by periods of growth (anagen), regression (catagen), and rest (telogen), after which time the hair is shed (also known as exogen). Approximately 90 percent of follicles on the scalp at any given time are in the active growth phase (anagen). During anagen, mitotically active matrix cells in the hair bulb differentiate and divide, resulting in a rate of hair growth of approximately 0.35 mm per day. Approximately 5 to 10 percent of follicles are in telogen (dormancy), during which all mitotic activity is arrested. The remaining 1 to 3 percent are in catagen, the involution phase. (See "Evaluation and diagnosis of hair loss", section on 'Hair cycle'.)

The final step of the hair cycle, exogen, is when the hair is released from the follicle. The scalp is estimated to contain on average 100,000 hairs, of which 100 to 150 are lost daily as part of the normal hair cycle. This loss typically occurs after washing and brushing the hair, so patients who wash their hair less frequently may note a greater number of hairs falling out at each instance.

Multiple signaling molecules have been implicated in the initial development and subsequent cycling of the hair follicle, including Wnt, sonic hedgehog, notch, and bone morphogenic proteins, among others [15]. In a mouse model of chemotherapy-induced alopecia, transient overexpression of sonic hedgehog accelerated hair fiber regrowth [16]. In feather follicles, chemotherapy damage to rapidly proliferating epithelial tissue has been shown to be mediated through downregulation of sonic hedgehog transcription; thus sonic hedgehog could conceivably be a future target for prevention of alopecia [17].

EFFECTS OF SYSTEMIC THERAPY

Pathophysiology — Cytotoxic chemotherapy attacks rapidly dividing cells in the body, including the dividing hair matrix cells [18]. This can result in alopecia by one or both of two mechanisms (figure 2):

If proliferation of the hair follicle matrix keratinocytes is severely inhibited, the hair may separate at the bulb and shed, a process referred to as anagen effluvium. Depending on the degree of toxicity on hair matrix keratinocytes, agents or schedules with lower toxicity will result in a dystrophic anagen effluvium, resulting in less alopecia and delayed hair regrowth; conversely, agents with greater toxicity will lead to severe alopecia but possibly more rapid hair regrowth [18]. (See "Evaluation and diagnosis of hair loss", section on 'Nonscarring alopecia'.)

Thinning of the hair shaft can occur at the time of maximal chemotherapy effect, resulting in Pohl-Pinkus constrictions. As a result, the hair shaft may break at the follicular orifice during the resting phase of the hair cycle. (See "Evaluation and diagnosis of hair loss", section on 'Trichoscopy'.)

Reversibility of alopecia is related to the degree of damage to the hair follicle stem cells [18]. Because chemotherapy effects are typically specific for proliferating cells, which reside in the bulb, sparing the quiescent stem cells in the bulge (figure 1) that are responsible for reinitiating follicle growth, alopecia from chemotherapy is usually, but not always, completely reversible. (See 'Recovery and reversibility' below.)

There are statistically significant differences between Asian, white population, and African American hair, relating to its growth rate, shaft shape and width, and mechanical properties [19].

Clinical characteristics — The term alopecia refers to the partial or complete absence of hair from any area of normal hair growth within the body. Chemotherapy-induced alopecia is most prominent on the scalp, with a predilection for areas with low total hair densities, in particular the crown and frontal areas of the scalp [20,21], where there is slower hair recovery. These are also areas most frequently affected by age-related alopecia and androgenetic alopecia, a process related to the effect of androgens on the hair follicle. Total scalp alopecia is most common, but alopecia can also be diffuse or patchy.

Loss of eyebrows and eyelashes (madarosis), extremity, as well as axillary and pubic hair, is variable, and may even occur after the last dose of chemotherapy has been administered. However, recovery is generally more rapid for hair in these areas than for hair on the scalp.

The timing of alopecia depends on the type(s) of systemic therapy agent, dose, and schedule. For most regimens that are given every two to three weeks, alopecia starts around two to three weeks and is completely lost by the end of the second cycle of chemotherapy. Weekly chemotherapy generally results in slower and occasionally incomplete alopecia, and hair may actually start to grow back with continuing treatment. High-dose chemotherapy used in the setting of hematopoietic cell transplantation leads to rapid and complete alopecia [22].

Some chemotherapy agents may cause prolonged or permanent alopecia, most notably docetaxel given at doses of 75 mg/m2 or higher per cycle, and less commonly paclitaxel [23-27].

In one prospective study of 61 patients treated for breast cancer, the proportion of participants who had prolonged chemotherapy-induced alopecia at six months and three years was 39.5 and 42.3 percent, respectively [24]. Most cases involved incomplete hair regrowth.

In another study of 100 patients with persistent chemotherapy-induced alopecia, the clinical presentation was most often hair thinning in the distribution of female pattern hair loss, and trichoscopic and histopathologic features indistinguishable from those of androgenetic alopecia [26]. Cosmetically significant regrowth was observed in a significant proportion of patients with topical or systemic treatments, suggesting at least partial reversibility. (See 'Interventions for chemotherapy-related alopecia' below.)

The effect of chemotherapy on hair regrowth is clearly dose and schedule related. It is important to advise patients about this risk before starting treatment with a specific regimen, as scalp cooling may ameliorate this risk. (See 'Recovery and reversibility' below.)

Chemotherapy, immune therapy, molecularly targeted, and endocrine agents may have effects on the hair other than alopecia:

Methotrexate and some targeted biologic agents may temporarily affect follicle melanocytes, inducing hyperpigmentation of scalp hair, eyebrow hair, and eyelashes; this tends to occur in bands that alternate with the normal color, a feature known as the "flag sign." This results from alternating periods of treatment and no treatment. (See "Cutaneous adverse effects of conventional chemotherapy agents".)

Small molecule inhibitors and monoclonal antibodies targeting epidermal growth factor receptor (EGFR), BRAF, Bruton tyrosine kinase (BTK), Bcr/Abl, cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), KIT, and platelet-derived growth factor receptor (PDGFR)/vascular endothelial growth factor receptor (VEGFR) may result in partial alopecia, hair curling, and dyspigmentation [28]. (See "Cutaneous adverse events of molecularly targeted therapy and other biologic agents used for cancer therapy", section on 'Other reactions'.)

Additional agents may cause partial (mild) alopecia, including targeted biologic agents, antibody-drug conjugates, and standard endocrine therapy (particularly tamoxifen and aromatase inhibitors) used in the adjuvant or metastatic setting [29]. Hair thinning with adjuvant endocrine therapy for early stage breast cancer has been associated with poor adherence with therapy [30]. (See 'Endocrine therapy' below.)

Although alopecia is relatively uncommon (1 to 2 percent), immunotherapy with immune checkpoint inhibitors may cause patchy hypopigmentation of the skin (vitiligo) and hair (poliosis), as well as alopecia [13,31]. (See "Cutaneous immune-related adverse events associated with immune checkpoint inhibitors", section on 'Hair and nail toxicities'.)

Quantitation — An alopecia grading scale for treatment-related alopecia is provided in the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) (table 1). In addition, the psychosocial impact of alopecia on patients may be qualified using the Patient-Reported Outcomes (PRO)-CTCAE scale (table 2) [32] and the Chemotherapy-induced Alopecia Distress Scale (CADS) [33]. A more detailed grading scale used to assess the effectiveness of alopecia prevention strategies was developed by Dean and is referred to as the Dean Scale (table 3) [34,35]. For objectively determining quantity of hair for research purposes, the Hair Mass Index can be applied [36].

Risk factors — The ability of chemotherapy agents to cause alopecia depends on the specific agent and the route, dose, and schedule of drug administration.

Risk differs substantially between chemotherapy agents, with a number of agents causing little to no alopecia (table 4).

High-dose, intermittent, intravenous chemotherapy regimens are associated with a high incidence of CTCAE grade 2 (complete or total, (table 1)) alopecia. Some targeted agents are also associated with grade 2 alopecia, especially the hedgehog signaling inhibitors and the fibroblast growth factor receptor inhibitors. (See 'Agents with highest risk' below.)

Low-dose therapy, oral administration, and weekly intravenous regimens are less likely to induce CTCAE grade 2 alopecia [3,4]. As an example, every-three-week, high- or moderate-dose, intravenous cyclophosphamide almost universally causes alopecia, while oral cyclophosphamide-containing regimens are less likely to do so. However, some weekly therapies cause grade 2 alopecia in most patients (eg, eribulin).

Combination chemotherapy regimens including drugs that cause at least some degree of alopecia are more likely to result in alopecia than single agents, depending, of course, on the agents and dose. The incidence of alopecia with common regimens has been inconsistently documented in the literature. (See "Overview of side effects of chemotherapy for early-stage breast cancer".)

A retrospective, case-control genome-wide association study (GWAS) of DNA samples from breast cancer patients treated with chemotherapy suggested that the single-nucleotide polymorphism rs3820706 in calcium channel voltage-dependent subunit beta 4 (CACNB4) on 2q23 (among others) was significantly associated with chemotherapy-induced complete alopecia (odds ratio 3.71, p = 8.13 x 10-9) [37]. This suggestion of individual risk assessment is intriguing but would require validation in a prospective trial given the very high rates of complete alopecia reported with standard chemotherapy for breast cancer.

Concomitant factors that can affect the risk and extent of chemotherapy-induced alopecia include poor drug metabolism (eg, patients with liver dysfunction may have unexpected, significant alopecia), prior exposure to scalp irradiation, older age, the presence of androgenic alopecia, use of prior chemotherapy causing alopecia, and the presence of graft-versus-host disease in those patients who have undergone hematopoietic cell transplantation [18,38,39]. By contrast, hair type, ethnicity, and race have not been associated with variations in either the extent of alopecia or the speed/pattern of hair regrowth. (See "Female pattern hair loss (androgenetic alopecia in females): Pathogenesis, clinical features, and diagnosis" and "Male pattern hair loss (androgenetic alopecia in males): Pathogenesis, clinical features, and diagnosis".)

Agents with highest risk

Cytotoxic agents — Of the commonly used intravenous single cytotoxic agents, those most likely to cause complete alopecia (dose and schedule dependent) include alkylating agents (cyclophosphamide, ifosfamide, busulfan, thiotepa), antitumor antibiotics (dactinomycin, doxorubicin, epirubicin, idarubicin), antimicrotubule agents (paclitaxel, docetaxel, ixabepilone, eribulin), and topoisomerase inhibitors (etoposide, irinotecan at higher doses) (table 4) [40]. Alopecia is less common or incomplete with bleomycin, low-dose epirubicin or doxorubicin (especially <30 mg/m2), oral cyclophosphamide, fluorouracil, capecitabine, gemcitabine, melphalan, methotrexate, mitomycin, mitoxantrone, the platinum agents (oxaliplatin, cisplatin, and carboplatin), topotecan, weekly low-dose irinotecan, and the vinca alkaloids (vinorelbine, vincristine, vinblastine).

Antibody drug conjugates (ADCs) represent highly effective treatments for various malignancies and cause variable rates of hair loss. As an example, the relatively new high drug-to-antibody ratio (DAR) ADCs (sacituzumab, govitecan, and fam-trastuzumab deruxtecan) and the lower DAR ADCs (enfortumab, vedotin) have been associated with variable degrees of hair loss (22 to 53 percent), whereas other ADCs (eg, trastuzumab emtansine) cause little to no hair loss. In general, the antibody part of the conjugate targets the specific cancer cell, and the conjugated toxin is released locally by proteolytic cleavage when the ADC is internalized by the cell. These next-generation ADCs also have membrane permeable toxins that leak out of the cancer cell, causing a so-called bystander effect, which enhances their efficacy. The degree of hair loss with ADCs is likely related to the specific toxin that is conjugated to each antibody, the DAR, the degree of bystander effect, and the quantity of free toxin that gets delivered to the hair follicle following lysis of the linker.

Endocrine therapy — CTCAE grade 1 alopecia is a common yet underreported adverse effect of estrogen antagonist therapies (such as tamoxifen) and aromatase inhibitors, is generally reversible with cessation of therapy, and is increased with endocrine therapies given in combination with targeted agents such as cyclin dependent kinase 4/6 inhibitors (CDK4/6i, see below) [29,41,42]. Rates of grade 1 alopecia for aromatase inhibitors have been reported in the range of 10 to 34 percent [29,43,44]. For tamoxifen, one meta-analysis concluded that the incidence of alopecia was 25 percent [41]. In this study, the mean time to develop alopecia from endocrine therapy initiation was 16.8 months (range 1 to 91 months) with 58 percent reporting alopecia within the first 12 months of therapy. The majority of affected patients taking either tamoxifen or aromatase inhibitors have androgenetic alopecia [42].

Molecularly targeted agents — Small molecule inhibitors of EGFR, as well as monoclonal antibodies targeting EGFR, can induce a constellation of cutaneous symptoms, which include an acneiform rash, abnormal hair growth (hirsutism), pruritus, and dry skin; together, this symptom complex is referred to by the acronym PRIDE (papulopustules and/or paronychia, regulatory abnormalities of hair growth, itching, dryness due to EGFR inhibitors). (See "Cutaneous adverse events of molecularly targeted therapy and other biologic agents used for cancer therapy", section on 'EGFR inhibitors'.)

In addition, diffuse or partial alopecia may occur with a number of targeted agents [28]:

The alopecia associated with agents that target EGFR is typically nonscarring and, therefore, reversible. However, anecdotal reports describe patients on long-term therapy who developed scarring alopecia [45,46], likely as a result of follicular and scalp skin infection. Interestingly, mice with a targeted deletion in EGFR gradually develop scarring alopecia [47].

Reversible alopecia is also described in patients receiving treatment with orally active multitargeted tyrosine kinase inhibitors (TKIs) (with wide variations in incidence), such as the KIT/PDGFRA inhibitor ripretinib (approximately 50 percent), VEGF inhibitors sorafenib (approximately 29 percent), regorafenib (approximately 24 percent), cabozantinib (approximately 16 percent), pazopanib (approximately 12 percent), axitinib (approximately 8 percent), sunitinib (approximately 7 percent), vandetanib (not reported); the BRAF inhibitors vemurafenib (approximately 24 percent), dabrafenib (approximately 19 percent), and encorafenib (approximately 14 percent); the Bcr/Abl inhibitors nilotinib (approximately 16 percent), dasatinib (approximately 8 percent), and imatinib (approximately 7 percent); and inhibitors of the fibroblast growth factor receptor (eg, pemigatinib) [28,48-53]. Interestingly, oral TKIs targeting HER2 do not cause significant alopecia.

Alopecia is reported in 57 percent of patients treated with vismodegib, an orally active agent approved for advanced basal cell cancer that inhibits sonic hedgehog signaling [54]. It is less common (approximately 49 percent) with the related agent sonidegib, also approved for advanced basal cell cancer [55], and it is even less common (approximately 30 percent) in patients treated with the related agent glasdegib, which is approved for acute leukemia in older adult patients [56]. (See "Systemic treatment of advanced basal cell and cutaneous squamous cell carcinomas not amenable to local therapies", section on 'Vismodegib'.)

CDK4/6i have demonstrated marked efficacy in combination with endocrine therapy in patients with metastatic hormone receptor-positive breast cancer, and abemaciclib has recently demonstrated efficacy in high-risk, early-stage disease. Three agents have regulatory approval in the metastatic setting: palbociclib, ribociclib, and abemaciclib. Abemaciclib in combination with endocrine therapy for two years in patients with high-risk, early-stage breast cancer improves disease-free and distant disease-free survival and has recently obtained regulatory approval in this setting. Given in combination with aromatase inhibitors or fulvestrant, CDK4/6i increase all-grade and CTCAE grade 2 alopecia compared with that observed with endocrine therapy alone. There is approximately a doubling of all-grade alopecia, from 10 to 16 percent to 25 to 33 percent, and approximately a 1.5 percent increased incidence of CTCAE grade 2 alopecia [43,44]. This alopecia appears to be reversible and may be treated with topical or oral minoxidil, with recent data suggesting efficacy from very low-dose oral minoxidil. (See 'Topical and oral minoxidil' below and "Treatment for hormone receptor-positive, HER2-negative advanced breast cancer", section on 'AIs plus CDK 4/6 inhibitors'.)

Recovery and reversibility — Because chemotherapy effects are typically specific for proliferating cells, which reside in the bulb, sparing the quiescent stem cells in the bulge that are responsible for reinitiating follicle growth, alopecia from chemotherapy is usually reversible. The hair follicle resumes normal cycling within a few weeks of treatment cessation, and visible regrowth becomes apparent within three to six months. The new hair frequently has different characteristics from the original; 65 percent of patients experience a graying, curling, or straightening effect, which is likely due to differential effects of chemotherapy on hair follicle melanocytes and inner root sheath epithelia, and these effects often resolve over time [3].

Although permanent alopecia is uncommon after standard-dose chemotherapy, there is now convincing evidence of permanent or prolonged alopecia after standard-dose chemotherapy for breast cancer (particularly with docetaxel and clearly related to both dose per infusion and duration of exposure) [24,27,57-60]. One study additionally described a genetic risk factor, in the ABCB1 gene, associated with permanent docetaxel-related alopecia in breast cancer patients [61]. There is one case series suggesting cases of delayed recovery with paclitaxel, although this is uncommon with current dosing schedules [23]. (See 'Anatomy and physiology' above.)

The impact of alopecia and potential alternative chemotherapy approaches should be discussed with each patient before the initiation of therapy that may lead to alopecia. This preemptive approach is important for minimizing the emotional distress associated with hair loss. For patients with breast cancer who are receiving docetaxel at doses of 75 mg/m2 or above per infusion, it is important to advise patients about the risk of prolonged or permanent alopecia. Scalp cooling may prevent permanent alopecia, although the data are limited [59]. (See 'Scalp hypothermia (scalp cooling)' below.)

PREVENTION OF ALOPECIA — Therapeutic approaches include physically decreasing the amount of drug delivered to the dividing hair bulb by reducing scalp blood flow, and pharmacologic or biologic interventions to block the effects of the chemotherapy on the hair follicle.

Scalp hypothermia (scalp cooling) — Scalp hypothermia can be offered to patients with a variety of solid tumors receiving bolus or short-term infusional chemotherapy associated with a moderate or high risk of alopecia. The evidence for benefit is most robust in patients treated for breast cancer; however, scalp hypothermia has been used successfully in patients with a variety of other solid tumors receiving chemotherapy regimens associated with a moderate to high risk of complete alopecia, including ovarian and prostate cancers. In addition, patients with advanced cancer for whom alopecia represents an unacceptable toxicity from palliative chemotherapy in the advanced stage setting may be offered the option of scalp hypothermia. (See 'Indications, efficacy and side effects' below.)

Two automated scalp cooling devices, the DigniCap and Paxman scalp hypothermia systems, are now US Food and Drug Administration (FDA) cleared based on two prospective clinical trials in patients receiving (neo)adjuvant chemotherapy for breast cancer. FDA clearance has been extended to cover patients with all solid tumors. Manual caps are also available, although there is a paucity of prospective controlled trials on the efficacy and safety of these caps in the United States. (See 'Mechanism of benefit, available devices, and technique' below.)

The decision to pursue scalp hypothermia must be individualized with shared decision-making that is based on risks and benefits, and the patient's values and preferences. Patients considering scalp hypothermia should be counseled on the variable efficacy, potential side effects, time requirements, and cost. Efficacy depends on the type and intensity of planned chemotherapy, with significantly less hair preservation in patients receiving anthracyclines compared with non-anthracycline-based regimens:

Despite scalp cooling, success in preventing alopecia in patients receiving the combination of anthracyclines with cyclophosphamide for breast cancer is modest, with up to 60 percent of patients still experiencing CTCAE grade 2 (≥50 percent hair loss) alopecia.

By contrast, the majority of patients receiving taxanes alone for breast cancer will retain the majority of their hair; >60 percent of patients receiving taxanes report no worse than CTCAE grade 1 (≤50 percent) alopecia.

Success also appears related to experience, cap fit, hair type and thickness, and likely issues related to drug exposure and clearance. These data, along with information about potential side effects (cold intolerance, headaches, forehead pain, lightheadedness), and financial burden and time requirements, should be discussed with each patient and are outlined in detail below. (See 'Indications, efficacy and side effects' below and 'Barriers to use, including reimbursement issues' below and 'Preparation protocol and special considerations for people of color' below.)

Not all patients should use scalp hypothermia, and contraindications are based on lack of efficacy in specific situations or concerns about safety in patients with various underlying diseases. Scalp hypothermia is not recommended for pediatric patients; adult patients with solid tumors receiving continuous infusion chemotherapy over ≥24 hours or those receiving brain radiotherapy; or patients with leukemia or some forms of lymphoma; and for those patients undergoing bone marrow or stem cell transplantation with myeloablative doses of chemotherapy and/or radiation therapy. Scalp hypothermia is contraindicated in patients with cold agglutinin disease, cryoglobulinemia, or post-traumatic cold dystrophy.

Mechanism of benefit, available devices, and technique — The mechanism of action of scalp hypothermia includes local vasoconstriction of blood vessels, resulting in reduced delivery of chemotherapy to the scalp, decreased follicle cell metabolic rate, and reduced cellular drug uptake [3,62-64].

Two primary approaches to scalp hypothermia are currently available: FDA-cleared automated systems that circulate coolant through cooling caps and maintain a constant temperature; and manual frozen gel caps (unregulated) that must be much colder than the automated system when applied to the scalp and are changed as they warm after approximately 30 minutes on the scalp (table 5). Automatic devices use a portable cooling unit that circulates a coolant in a flexible cap so that temperature is maintained within a narrow range. Cooling with manual gel caps requires a cooler with dry ice or a freezer (if available in the chemotherapy infusion center).

For the frozen gel caps, generally patients must arrange to freeze the caps ahead of their infusion, then transport and store them in a cooler with dry ice. In addition, there must be someone accompanying the patient who can change the cap every 25 to 30 minutes, depending on the method of freezing. These gel caps are generally stored in the refrigerator until they need to be frozen again before the next chemotherapy session.

Regardless of the specific device that is used, scalp cooling is started approximately 30 minutes before the chemotherapy infusion starts in order to allow gradual cooling of the scalp to the desired temperature. The duration of postinfusion cooling is determined at least in part by the clearance of high levels of chemotherapy, but also by the severity of the expected alopecia. Post-infusion cooling times are highly variable, and at least to some degree are institution and manufacturer dependent. For many centers, a 90-minute post-cooling time is used for weekly paclitaxel, with longer times for other chemotherapy agents (and combinations), including every-three-week docetaxel, with cooling maintained for up to three hours after the end of the chemotherapy infusion [65]. However, some studies have suggested that a post-infusion cooling time of 20 minutes might be sufficient for single-agent docetaxel and weekly paclitaxel, with success in preventing hair loss that is similar to a longer duration of 45 minutes [66-68]. It is important to note that these data are specifically in patients receiving single-agent taxane therapy.

Generally, an insulating cap is placed over the cold cap, and a protective covering is placed over the head between the scalp and cold cap in places where the cap may come in direct contact with unprotected skin (eg, forehead, top of the ears). For most devices, caps are available in various sizes and fitted to the patient's specific head size in advance. One device from Dignitana ("Delta") provides a cap that can be shaped to the individual head (similar to the design of the frozen Penguin cap, but with an automated circulating coolant).

The optimal scalp skin temperature for successful cooling is thought to be ≤18ºC [69], although data regarding this specific number are quite limited, and it is difficult to accurately measure the actual temperature of the scalp. However, the caps themselves must be much colder in order to bring the scalp to the desired temperature. The type of cooling device determines the temperature, as manual caps must be much colder when initially applied to the scalp to account for their gradual increase in temperature before a new cap is applied. The automated units are usually set around 0°C.

Indications, efficacy and side effects — Scalp hypothermia is an option for patients with a variety of solid tumors receiving bolus or short-term infusional chemotherapy with a high risk of alopecia. The evidence for benefit is most robust in patients treated for breast cancer, especially with taxanes. However, scalp hypothermia has been used successfully in patients with a variety of other solid tumors receiving chemotherapy regimens associated with a high risk of complete alopecia, including ovarian and prostate cancers. In addition, patients with advanced cancer for whom alopecia represents an unacceptable toxicity from palliative chemotherapy in the advanced stage setting may be offered the option of scalp hypothermia.

Scalp hypothermia is now widely used both within and outside of the United States to prevent or reduce chemotherapy-induced alopecia. Scalp cooling is listed as an option for alopecia prevention in the National Comprehensive Cancer Network (NCCN) guidelines for breast cancer [70] and has been incorporated in the European Society of Medical Oncology (ESMO) Clinical Practice Guidelines [71].

Reported success has been variable, depending on the specific cooling device and type of chemotherapy [72,73]. In general, cooling has been less effective when used with combination regimens that include an anthracycline, although this is somewhat dependent on dose and schedule, and to some degree, experience in the optimal use of caps [38,39,74-77].

The following studies have addressed efficacy and safety:

A meta-analysis in 2015 concluded that scalp hypothermia was the only intervention that significantly reduced the risk of chemotherapy-induced alopecia (10 studies, involving 818 patients and including three randomized trials; relative risk 0.38 compared with no cooling, 95% CI 0.32-0.45) [73]. A subgroup analysis suggested a similar degree of efficacy for scalp hypothermia regardless of the underlying cancer, but the majority of patients included in the analysis had breast cancer, and most of the trials that included other patients did not provide detailed diagnoses. No significant adverse events associated with scalp hypothermia were reported in this meta-analysis, although the reported studies did not always track toxicity.

In a later meta-analysis of 27 studies (three randomized trials, 12 cohort, and 12 cross-sectional studies) limited to breast cancer patients undergoing chemotherapy who used a variety of scalp cooling devices, estimates of adverse reactions that were reported in four or more studies included headache (28 percent), scalp pain (25 percent), feeling cold (50 percent), neck pain (22 percent), and heaviness of the head (53 percent) [78].

Both the benefits of and minimal to modest adverse events from scalp cooling have been confirmed in at least three contemporary prospective trials evaluating the efficacy of two scalp hypothermia devices in women with early stage breast cancer [79-82]:

In one multicenter, prospective cohort study, 101 patients with early stage breast cancer receiving non-anthracycline taxane-based chemotherapy who used the DigniCap scalp cooling device were compared with 16 concurrently treated controls who did not use the cooling device [79]. Alopecia was measured using the Dean Scale, with success defined as alopecia of 50 percent or less (Dean score 0 to 2 (table 3)) one month after the last chemotherapy infusion, and was graded by the patients themselves using photographs compared with their own baseline hair.

The rate of significant alopecia (>2 on the Dean Scale) was 50 percent or less in 66.3 percent of the intervention group compared with none of the control group (p <0.001). Three of five quality of life measures were significantly better one month after the end of chemotherapy, including perception of hair loss, feeling upset over hair loss, and feeling less physically attractive as a result of the disease or its treatment. The primary toxicity was mild headache, and three patients stopped cooling due to feeling cold.

A second trial randomly assigned 182 patients in a 2 to 1 ratio to use of the Paxman scalp cooling device or no scalp hypothermia during chemotherapy for breast cancer; 36 percent received anthracycline-based chemotherapy, while the remainder received a taxane, either alone or in combination with carboplatin, cyclophosphamide, pertuzumab, and/or trastuzumab [80]. Successful hair preservation was defined as less than 50 percent hair loss not requiring a wig, using the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) criteria (grade 0 to 1 (table 1)), and was graded by a clinician unaware of treatment assignment at the end of four cycles of chemotherapy.

The interim analysis included 142 participants evaluable for the alopecia endpoint; all had completed four cycles of their assigned chemotherapy. Scalp hypothermia was graded as successful in 50.5 percent of patients compared with none of the control group (p = 0.0061). Adverse events were all grade 1 and 2, including primarily headache and feeling cold. Interestingly, in this study, there were substantial differences in the success of cooling by site and by drug group. An exploratory post hoc analysis indicated that only 16 percent of patients receiving anthracycline-based chemotherapy met criteria for success, compared with 59 percent of those receiving taxanes, although the confidence intervals were very wide. Seven patients discontinued cooling early, primarily due to feeling cold.

A third study evaluated the use of DigniCap in 139 patients with early stage breast cancer receiving adjuvant chemotherapy for their breast cancer [81]. The majority (95 percent) received at least four cycles of anthracycline-based chemotherapy (eribulin and cyclophosphamide), and many then received sequential taxanes. Using the Dean Scale (with success defined as a self-assessed score of <2), three weeks after completing chemotherapy, 131 patients were evaluable for efficacy. Overall, 56 of the 131 patients reported success, for a rate of 43 percent. When evaluating the 104 patients who completed scalp cooling with all chemotherapy sessions, the success rate was higher at 54 percent. Overall, cooling was well tolerated, with nine patients discontinuing treatment due to adverse events.

Although this study did meet its endpoint of success in 55 percent of patients, 43 percent of patients lost less than 50 percent of their hair, which may be acceptable to some patients. It is hoped that better cap designs with the automated devices will improve the success with scalp cooling in patients treated with combination anthracyclines.

Adverse events from scalp hypothermia are generally mild and include patient discomfort from feeling cold, headache, nausea, dry skin, and claustrophobia. The manual caps have been reported to cause scalp thermal injury, which can probably be avoided by using an inner protective cap or a band at the edge of the hair [79,80,83-88]. It has been suggested that use of scalp hypothermia may result in faster regrowth of hair following completion or chemotherapy, and in some cases regrowth has been reported during chemotherapy with continued use of scalp cooling [89-93]. Although this is not well documented, certainly retaining some hair at the end of chemotherapy could simply be associated with a shorter time to having an "acceptable" density of hair. A retrospective study in Spain including 492 patients who received docetaxel as adjuvant therapy for early stage breast cancer observed that fewer patients who had scalp cooling during chemotherapy developed persistent chemotherapy-induced alopecia when compared with uncooled contemporary and historical controls [59].

Is there a risk for scalp metastases? — Previously, some investigators have raised concerns regarding the possibility of increasing the risk of scalp skin metastasis in patients who have used scalp hypothermia during chemotherapy [3,4,62,63,83,94-98]. This concern is based on the hypothesis that stem cells or malignant cells that may be present in the scalp at the time of treatment are not adequately treated when the scalp is cooled due to reduced delivery of drug to the scalp.

Breast cancer – The incidence of scalp skin metastasis in patients who have used scalp hypothermia devices has been best studied in breast cancer, where the overall risk of scalp metastasis is very low, and these are often discovered either along with or following a diagnosis of systemic metastatic disease:

In one study of 61 patients with breast cancer receiving chemotherapy who used cooling caps, only one patient with underlying liver dysfunction developed cutaneous scalp metastasis [62].

Larger studies evaluating patients using scalp hypothermia during chemotherapy for early-stage breast cancer have shown no association between use of a cooling device and the subsequent development of scalp skin or skull metastases, and one large study showed no impact of scalp hypothermia on survival [97,99,100]. In one review, the incidence of scalp skin metastasis in breast cancer patients was comparable for scalp-cooled (0.04 to 1 percent) and non-scalp-cooled patients (0.03 to 3 percent) [99].

Additional information is available from a systematic review and meta-analysis of 1959 patients using scalp cooling devices over an estimated mean time frame of 43.1 months and 1238 patients who did not use a scalp cooling device over an estimated mean time frame of 87.4 months [97]. The incidence rate of scalp metastasis in the scalp cooling group versus the no scalp cooling group was 0.61 (95% CI 0.32-1.1 percent) versus 0.41 percent (95% CI 0.13-0.94 percent); p = 0.43.

In the prospective DigniCap study described above, no patient has developed scalp metastases at a median follow-up of four years [79]. (See 'Indications, efficacy and side effects' above.)

Other cancers – Data on the risk of scalp metastases with scalp hypothermia for other tumors is extremely limited. Scalp cooling is not used in patients with hematologic malignancies treated for curative intent, including leukemia and some forms of lymphoma because of the circulating nature of the malignant cells [96,98,101].

Other contraindications — Scalp hypothermia is contraindicated in several other situations [101]:

Scalp cooling caps are not designed for pediatric patients, so scalp cooling is not recommended in this setting.

Patients with solid tumors receiving continuous-infusion chemotherapy regimens over one day or longer that result in alopecia, and those undergoing whole-brain or targeted brain irradiation are also not good candidates for scalp hypothermia due to the ineffectiveness of cooling in these situations [96,98,102].

Scalp hypothermia is contraindicated in patients with cold agglutinin disease, cryoglobulinemia, and post-traumatic cold dystrophy due to potential toxicity [72].

Scalp hypothermia may be less effective in patients with significant liver dysfunction due to delayed drug metabolism, thereby allowing persistence of therapeutic drug levels for a long duration of time and increasing the risk of alopecia.

Barriers to use, including reimbursement issues — Barriers to use of scalp hypothermia were addressed in a survey of 155 providers through the ASCO Research Survey Pool [103]. Providers who treated breast cancer, those who were familiar with scalp hypothermia and read related literature in the past two years, and those who had access to machine scalp hypothermia were more likely to discuss this option with patients. Financial concerns were the primary reason for not recommending scalp hypothermia, and a small percentage of providers still had safety concerns, despite continued data about safety in terms of cancer risk. Clearly familiarity and experience lead to greater support and utilization of scalp hypothermia [104].

The cost of using scalp cooling varies depending on the number of cycles of chemotherapy and the type of cooling device used, but the average total cost for scalp hypothermia is estimated to range between USD $1500 and $3000 per patient depending on the number of treatment cycles [104]. In addition, there may be institutional costs associated with extra time in the chemotherapy infusion center and additional personnel costs.

In the United States, The US Centers for Medicare and Medicaid Services (CMS) has reassigned payment for scalp hypothermia for Medicare claims filed using current procedural terminology (CPT) code 0662T. The treatment has been reassigned to New Technology APC 1520 with a national average payment of $1850.50, effective January 1, 2022. Some private health insurance companies also provide partial coverage, although this is highly variable. Because of the variable and mostly absent coverage until 2022, use of scalp hypothermia may be associated with financial concerns and variable costs for patients. One philanthropic organization provides some financial assistance to those who cannot afford scalp hypothermia (hairtostay.org), and it is hoped that private insurance coverage will improve with clear efficacy and safety data as well as the change in Medicare coverage, although some policies specifically exclude any procedure for "cosmetic" purposes and have used this as a reason not to cover scalp cooling with chemotherapy [104,105]. In most European countries, scalp cooling is covered by the CPT code, so there are no additional costs for patients.

Preparation protocol and special considerations for people of color — Although there is no standard preparation protocol for scalp cooling, patients are typically asked to wet the hair or apply conditioner before donning the cap to improve conductivity and reduce pretreatment cooling time [106]. Areas of the scalp with inadequate contact with the cap are not protected from alopecia [72].

The effectiveness of scalp cooling may also be affected by hair type, thickness, and prior hair treatments, especially in people of color [107-109]:

Black kinky or curly hair has different physical properties as compared with straight hair, especially when it is wet. While straight hair flattens when wet, kinky or curly hair tends to become more kinky/curly.

The physical properties of curly hair are often altered in response to heat and chemical treatments (such as a permanent wave or chemical straightening) to a much greater extent than straight hair, assuming a shape or form that varies under different conditions [110]. As an example, when curly hair is straightened using heat, it appears flat and straight when dry, but returns to its curly state when dampened. On the other hand, kinky or curly hair that has been straightened using chemicals remains straight regardless of being dry or wet.

Another factor is that people who have tightly curled hair often wear braids and weaves that involve hair extensions to protect hair from the damage of daily grooming; these may interfere with cap-scalp contact. In addition, the use of scalp cooling in patients with braids and exposed scalp in between the braids could increase the potential risk for cold related skin injury, so special care should be taken along with careful observation.

Suggestions to enhance the likelihood of treatment success for scalp hypothermia in people with kinky or curly hair are outlined in the table (table 6) [108].

Interventions for chemotherapy-related alopecia — Preclinical studies suggest that both small molecules and biologic agents may reduce or prevent alopecia by protecting the hair bulb from the damaging effects of chemotherapy. The interventions that have been specifically tested in humans include topical and oral minoxidil, topical bimatoprost, oral finasteride, spironolactone, and topical calcitriol. At present, there are no pharmacologic interventions that have been approved by regulatory agencies for this indication.

Topical and oral minoxidil — For patients with delayed recovery of chemotherapy-induced alopecia or endocrine therapy-associated hair loss, we suggest a trial of minoxidil. For most patients with chemotherapy-induced alopecia or significant alopecia related to endocrine therapy (with or without targeted agents), we suggest oral rather than topical minoxidil; for those with less significant hair loss related to endocrine therapy, either oral or topical minoxidil is appropriate. Minoxidil should be used only for treatment, not prevention of chemotherapy-related alopecia.

Minoxidil stimulates ATP-sensitive potassium channels, resulting in vasodilation and premature entry of resting hair follicles into the anagen or growth phase. It may also increase hair follicle size, thereby counteracting miniaturization of the hair follicle, which is the characteristic histologic finding of androgenetic alopecia.

Topical – Topical minoxidil has been FDA approved for the treatment of androgenetic alopecia. (See "Male pattern hair loss (androgenetic alopecia in males): Pathogenesis, clinical features, and diagnosis", section on 'Pathogenesis' and "Female pattern hair loss (androgenetic alopecia in females): Pathogenesis, clinical features, and diagnosis" and "Female pattern hair loss (androgenetic alopecia in females): Management" and "Male pattern hair loss (androgenetic alopecia in males): Management".)

It has also been successfully used for the treatment of endocrine therapy-related alopecia in patients with breast cancer, although the available data are limited, and many patients find use difficult to maintain.

The best data come from a series of 112 patients with alopecia related to aromatase inhibitors (67 percent) or tamoxifen (33 percent) who had not received prior cytotoxic chemotherapy [42]. In 96 of the 102 patients with standardized clinical photographs, alopecia was CTCAE grade 1 (table 1). Response to topical minoxidil was assessed using standardized clinical photographs in 46 of the 112 patients. Overall, 37 (80 percent) showed a moderate to significant improvement in alopecia.

On the other hand, two randomized trials suggest that the effects of topical minoxidil in preventing or treating chemotherapy-induced alopecia are limited at best:

In a randomized trial of 48 patients with varying solid tumors receiving doxorubicin-containing regimens, topical minoxidil (2 percent solution applied twice daily) did not prevent the development of severe alopecia compared with placebo [111].

A second trial in 22 women receiving chemotherapy after surgery for breast cancer also found that treatment with topical minoxidil did not prevent alopecia, but it did shorten the time to maximal regrowth and the time from maximal alopecia to first regrowth, and lengthened the time to maximal alopecia [112].

Oral – Oral minoxidil appears to be effective in treating delayed hair recovery following systemic chemotherapy or alopecia resulting from endocrine therapy and is generally well tolerated.

One trial evaluated oral minoxidil (1.25 mg daily) in 216 patients with late alopecia, which was defined as incomplete hair regrowth ≥6 months following completion of cytotoxic chemotherapy alone (n = 31) or alopecia after initiation of oral endocrine therapy (n = 65) or alopecia following combined chemotherapy and endocrine therapy (n =120) [113]. A wide variety of chemotherapy agents had been used, and the majority of patients (79 percent) had a history of breast cancer. In a preliminary report presented at the 2022 annual ASCO meeting, clinical improvement as assessed by standardized photography was noted in 74 percent of patients. Among the 42 patients undergoing trichoscopy at a median of 91 days from the start of minoxidil, hair density improved by about 21 percent. No patient discontinued oral minoxidil because of adverse effects. A 2.5 mg dose has also been used in clinical practice without prospective data.

Combination of topical and oral – Limited data suggest benefit in regrowth with the combination of oral and topical minoxidil relative to topical minoxidil alone, but further data are needed. In a retrospective cohort study in 56 patients with breast cancer and therapy-induced alopecia, concomitant treatment with 1.25 to 5 mg daily of low dose oral minoxidil and once daily 5% topical minoxidil solution was associated with more frequent treatment responses compared with topical minoxidil (89 versus 53 percent) [114]; the combination was also associated with a higher percentage increase in hair density/cm2 from baseline (16 versus 6 percent). Post-treatment changes in hair thickness (micrometer) were similar between the two groups.

Topical bimatoprost — A trial of topical bimatoprost is reasonable for the treatment of chemotherapy-induced eyelash or eyebrow hypotrichosis, but there are no data supporting a preventive benefit, and safety during chemotherapy has not been evaluated.

Topical 0.03% bimatoprost, a prostaglandin analog, has been used successfully on the upper eyelid margin to enhance eyelash growth in patients with eyelash hypotrichosis. (See "Alopecia areata: Management", section on 'Eyelash loss'.)

Benefit for patients with chemotherapy-induced eyelid hypotrichosis was observed in a randomized controlled trial of 130 patients with idiopathic or chemotherapy-induced alopecia [115]. Eligible patients had completed chemotherapy within 4 to 16 weeks with documented eyelash hypotrichosis, and applied one drop to the upper eyelid margin of each eye once daily. The primary endpoint of at least a one-grade improvement in investigator-assessed Global Eyelash Assessment (GEA) and at least a three-point improvement in patient-reported Eyelash Satisfaction Questionnaire (ESQ) domain 2 at month 4 was met in the chemotherapy group, with significantly less recovery in the control group (37.5 percent for bimatoprost versus 18.2 percent for vehicle), and benefits were more pronounced at month 12. Bimatoprost can also be painted onto the eyebrows to improve hair growth at that site.

Finasteride — The data are insufficient to support use of finasteride for the treatment or prevention or treatment of alopecia in patients receiving chemotherapy, or in women with breast cancer and endocrine therapy-associated alopecia.

Finasteride, a type II 5-alpha-reductase inhibitor, is approved for the treatment of benign prostatic hyperplasia to improve symptoms, to reduce the risk of acute urinary retention or the need for surgical procedures, and to treat male pattern hair loss. There are limited data on its efficacy in women with female pattern alopecia, with variable success. (See "Male pattern hair loss (androgenetic alopecia in males): Management" and "Female pattern hair loss (androgenetic alopecia in females): Management".)

However, finasteride has been shown to increase serum estrogen levels in 34 percent of patients [116] and has been associated with gynecomastia in men. In our view, the potential hazards of finasteride outweigh any potential benefit.

Spironolactone — The data with spironolactone are limited, but this is a relatively nontoxic therapy that can be considered for treatment of persistent alopecia following cancer therapy.

Spironolactone is an aldosterone agonist that competitively blocks androgen receptors and weakly blocks androgen synthesis. It has been used to treat women with androgenetic alopecia, with limited but clear efficacy in some patients. (See "Female pattern hair loss (androgenetic alopecia in females): Management".)

There are very limited data on the benefit and safety of spironolactone either as a single agent or in combination with topical minoxidil in women with alopecia after chemotherapy or endocrine therapy [23,27]. Concern has been raised about the potential for increased estrogen levels and possibly higher rates of breast cancer with spironolactone, but this has not been consistently shown in patients with no history of the disease [116,117].

Topical calcitriol — The data are insufficient to support use of topical calcitriol for prevention of chemotherapy-induced alopecia.

Pretreatment with topical calcitriol (1,25(OH)2D3; the most active metabolite of vitamin D) protects rats from cyclophosphamide-, etoposide-, and doxorubicin-induced alopecia [118]. Effects may be mediated by direct biological activity or modulation of other factors. Specific receptors for calcitriol are present in rat, murine, and human skin cells, and calcitriol induces differentiation of murine epidermal keratinocytes. When human cultured keratinocytes are incubated with calcitriol, there is a dose- and time-dependent stimulation of differentiation and inhibition of DNA synthesis.

One study found that pretreatment with calcitriol did not alter the cytotoxic effects of the chemotherapy but it did prevent significant alopecia [119]. However, a phase I trial of 12 patients receiving anthracycline- and cyclophosphamide-containing chemotherapy for breast cancer failed to demonstrate any benefit in preventing chemotherapy-induced alopecia [120]. Furthermore, concerns about potential protection of the cancer cells from the effects of chemotherapy have been raised [118,119,121].

Photobiomodulation therapy — Limited data suggest accelerated hair regrowth with photobiomodulation therapy after chemotherapy in breast cancer patients [122]. A small randomized controlled trial (n = 32) in patients on anthracycline and taxane-containing chemotherapy showed significant higher numerical rating scale (NRS) scores for regrowth in the group treated with photobiomodulation at one month post-chemotherapy. In the control group NRS scores remained constant.

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, "The Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on "patient info" and the keyword(s) of interest.)

Basics topic (see "Patient education: Hair loss from cancer treatment (The Basics)")

SUMMARY AND RECOMMENDATIONS

Clinical features and risk factors – Alopecia is a consequence of systemic cancer therapy that can be psychologically devastating. Alopecia is generally most prominent on the scalp, and there is a predilection for areas that show low total hair densities, in particular the crown and frontal scalp. (See 'Clinical characteristics' above.)

The frequency and severity vary depending on the specific chemotherapy agent or combination regimen administered (table 4), drug dose, and the treatment schedule. The majority of cases are transient and reversible once therapy is discontinued, with the possible exception of higher dose docetaxel and some molecularly targeted therapies. (See 'Risk factors' above and 'Recovery and reversibility' above.)

All patients who will receive chemotherapy that may result in alopecia should be informed of this side effect and, for patients treated with docetaxel, the possibility of prolonged or permanent alopecia. Options such as scalp hypothermia, head wraps, hats, or wigs should be discussed in advance so that the patient can be physically and emotionally prepared to cope with this well described toxicity. (See 'Clinical characteristics' above and 'Recovery and reversibility' above.)

Scalp hypothermia – Scalp hypothermia minimizes delivery of chemotherapeutic agents to the scalp and reduces metabolism of hair follicle cells, thereby decreasing the risk of alopecia. (See 'Scalp hypothermia (scalp cooling)' above.)

Indications, types, and counseling

-Scalp hypothermia can be offered to patients with a variety of solid tumors receiving bolus or short-term infusional chemotherapy with a high risk of alopecia. The evidence for benefit is most robust in those treated for breast cancer; however, scalp hypothermia has been used successfully in patients with a variety of other solid tumors receiving chemotherapy regimens associated with a high risk of complete alopecia. (See 'Indications, efficacy and side effects' above.)

-Two automated scalp cooling devices, the DigniCap and Paxman scalp hypothermia systems, are now US Food and Drug Administration (FDA) cleared based on two prospective clinical trials in patients receiving (neo)adjuvant chemotherapy for breast cancer. FDA clearance has been extended to cover patients with all solid tumors. Manual caps are also available, although there is a paucity of prospective controlled trials on the efficacy and safety of these caps in the United States. (See 'Mechanism of benefit, available devices, and technique' above.)

-The decision to pursue scalp hypothermia must be individualized with shared decision-making that is based on risks and benefits, and the patient's values and preferences. Patients considering scalp hypothermia should be counseled on the variable success of this approach, particularly with anthracycline-based combination therapy, as well as potential adverse events of cold intolerance, headaches, forehead pain, and lightheadedness. (See 'Indications, efficacy and side effects' above.)

In addition, there is a financial burden of scalp hypothermia that should be discussed with each patient. (See 'Barriers to use, including reimbursement issues' above.)

Contraindications

-Scalp hypothermia is not recommended for pediatric patients due to the lack of pediatric-sized caps, or for adult patients with diseases associated with high levels of circulating tumor cells, such as leukemia and some types of lymphoma. In general, these patients are also receiving high doses of systemic chemotherapy where scalp cooling is unlikely to be effective.

-Scalp hypothermia is contraindicated or is unlikely to be effective in patients receiving radiation therapy to the brain or high doses of chemotherapy with bone marrow or stem cell rescue.

-Scalp hypothermia is contraindicated in patients with symptomatic cold agglutinin disease, cryoglobulinemia, and post-traumatic cold dystrophy.

Pharmacologic preventive and treatment strategies

For patients with delayed recovery of chemotherapy-induced alopecia or endocrine therapy-associated hair loss, we suggest minoxidil (Grade 2C). For most patients with chemotherapy-induced or significant endocrine therapy (with or without targeted agents) induced alopecia, we suggest low-dose oral rather than topical minoxidil (Grade 2C); for those with mild alopecia related to endocrine therapy, either oral or topical minoxidil is appropriate. (See 'Topical and oral minoxidil' above.)

A trial of topical bimatoprost is reasonable for the treatment of chemotherapy-induced eyelash alopecia, but there are no data supporting a preventive benefit, and safety during chemotherapy has not been evaluated. (See 'Topical bimatoprost' above.)

The available data are insufficient to support use of finasteride or topical calcitriol for prevention or treatment of cancer treatment-induced alopecia. (See 'Finasteride' above and 'Topical calcitriol' above.)

The data with spironolactone are limited, but this is a nontoxic therapy that can be considered for treatment of persistent alopecia following cancer therapy.

ACKNOWLEDGMENT — The editorial staff at UpToDate acknowledge Aimee Payne, MD, PhD, who contributed to earlier versions of this topic review.

  1. Choi EK, Kim IR, Chang O, et al. Impact of chemotherapy-induced alopecia distress on body image, psychosocial well-being, and depression in breast cancer patients. Psychooncology 2014; 23:1103.
  2. Gandhi M, Oishi K, Zubal B, Lacouture ME. Unanticipated toxicities from anticancer therapies: survivors' perspectives. Support Care Cancer 2010; 18:1461.
  3. Dorr VJ. A practitioner's guide to cancer-related alopecia. Semin Oncol 1998; 25:562.
  4. Hussein AM. Chemotherapy-induced alopecia: new developments. South Med J 1993; 86:489.
  5. McGarvey EL, Baum LD, Pinkerton RC, Rogers LM. Psychological sequelae and alopecia among women with cancer. Cancer Pract 2001; 9:283.
  6. Mulders M, Vingerhoets A, Breed W. The impact of cancer and chemotherapy: perceptual similarities and differences between cancer patients, nurses and physicians. Eur J Oncol Nurs 2008; 12:97.
  7. Lemieux J, Maunsell E, Provencher L. Chemotherapy-induced alopecia and effects on quality of life among women with breast cancer: a literature review. Psychooncology 2008; 17:317.
  8. Hesketh PJ, Batchelor D, Golant M, et al. Chemotherapy-induced alopecia: psychosocial impact and therapeutic approaches. Support Care Cancer 2004; 12:543.
  9. van den Hurk CJ, Mols F, Vingerhoets AJ, Breed WP. Impact of alopecia and scalp cooling on the well-being of breast cancer patients. Psychooncology 2010; 19:701.
  10. Trüeb RM. Chemotherapy-induced hair loss. Skin Therapy Lett 2010; 15:5.
  11. Elder D, Elenitsas R, Johnson BL, Murphy GF. Lever's Histopathology of the Skin, 9th ed, Lippincott Williams & Wilkins, Philadelphia 2004. p.1229.
  12. Paus R, Cotsarelis G. The biology of hair follicles. N Engl J Med 1999; 341:491.
  13. Zarbo A, Belum VR, Sibaud V, et al. Immune-related alopecia (areata and universalis) in cancer patients receiving immune checkpoint inhibitors. Br J Dermatol 2017; 176:1649.
  14. Liu Y, Lyle S, Yang Z, Cotsarelis G. Keratin 15 promoter targets putative epithelial stem cells in the hair follicle bulge. J Invest Dermatol 2003; 121:963.
  15. Cotsarelis G, Millar SE. Towards a molecular understanding of hair loss and its treatment. Trends Mol Med 2001; 7:293.
  16. Sato N, Leopold PL, Crystal RG. Effect of adenovirus-mediated expression of Sonic hedgehog gene on hair regrowth in mice with chemotherapy-induced alopecia. J Natl Cancer Inst 2001; 93:1858.
  17. Xie G, Wang H, Yan Z, et al. Testing chemotherapeutic agents in the feather follicle identifies a selective blockade of cell proliferation and a key role for sonic hedgehog signaling in chemotherapy-induced tissue damage. J Invest Dermatol 2015; 135:690.
  18. Paus R, Haslam IS, Sharov AA, Botchkarev VA. Pathobiology of chemotherapy-induced hair loss. Lancet Oncol 2013; 14:e50.
  19. Westgate GE, Ginger RS, Green MR. The biology and genetics of curly hair. Exp Dermatol 2017; 26:483.
  20. Chon SY, Champion RW, Geddes ER, Rashid RM. Chemotherapy-induced alopecia. J Am Acad Dermatol 2012; 67:e37.
  21. Yun SJ, Kim SJ. Hair loss pattern due to chemotherapy-induced anagen effluvium: a cross-sectional observation. Dermatology 2007; 215:36.
  22. Kanti V, Nuwayhid R, Lindner J, et al. Analysis of quantitative changes in hair growth during treatment with chemotherapy or tamoxifen in patients with breast cancer: a cohort study. Br J Dermatol 2014; 170:643.
  23. Freites-Martinez A, Chan D, Sibaud V, et al. Assessment of Quality of Life and Treatment Outcomes of Patients With Persistent Postchemotherapy Alopecia. JAMA Dermatol 2019; 155:724.
  24. Kang D, Kim IR, Choi EK, et al. Permanent Chemotherapy-Induced Alopecia in Patients with Breast Cancer: A 3-Year Prospective Cohort Study. Oncologist 2019; 24:414.
  25. Chan J, Adderley H, Alameddine M, 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 (Engl) 2021; 30:e13395.
  26. Bhoyrul B, Asfour L, Lutz G, et al. Clinicopathologic Characteristics and Response to Treatment of Persistent Chemotherapy-Induced Alopecia in Breast Cancer Survivors. JAMA Dermatol 2021; 157:1335.
  27. Kluger N, Jacot W, Frouin E, et al. Permanent scalp alopecia related to breast cancer chemotherapy by sequential fluorouracil/epirubicin/cyclophosphamide (FEC) and docetaxel: a prospective study of 20 patients. Ann Oncol 2012; 23:2879.
  28. Belum VR, Marulanda K, Ensslin C, et al. Alopecia in patients treated with molecularly targeted anticancer therapies. Ann Oncol 2015; 26:2496.
  29. Gallicchio L, Calhoun C, Helzlsouer KJ. Aromatase inhibitor therapy and hair loss among breast cancer survivors. Breast Cancer Res Treat 2013; 142:435.
  30. Moscetti L, Agnese Fabbri M, Sperduti I, et al. Adjuvant aromatase inhibitor therapy in early breast cancer: what factors lead patients to discontinue treatment? Tumori 2015; 101:469.
  31. Burzi L, Alessandrini AM, Quaglino P, et al. Cutaneous Events Associated with Immunotherapy of Melanoma: A Review. J Clin Med 2021; 10.
  32. Kluetz PG, Chingos DT, Basch EM, Mitchell SA. Patient-Reported Outcomes in Cancer Clinical Trials: Measuring Symptomatic Adverse Events With the National Cancer Institute's Patient-Reported Outcomes Version of the Common Terminology Criteria for Adverse Events (PRO-CTCAE). Am Soc Clin Oncol Educ Book 2016; 35:67.
  33. Cho J, Choi EK, Kim IR, et al. Development and validation of Chemotherapy-induced Alopecia Distress Scale (CADS) for breast cancer patients. Ann Oncol 2014; 25:346.
  34. Dean JC, Salmon SE, Griffith KS. Prevention of doxorubicin-induced hair loss with scalp hypothermia. N Engl J Med 1979; 301:1427.
  35. Messenger AG, Sinclair R. Follicular miniaturization in female pattern hair loss: clinicopathological correlations. Br J Dermatol 2006; 155:926.
  36. Komen MMC, van den Hurk CJG, Nortier JWR, et al. Patient-reported outcome assessment and objective evaluation of chemotherapy-induced alopecia. Eur J Oncol Nurs 2018; 33:49.
  37. Chung S, Low SK, Zembutsu H, et al. A genome-wide association study of chemotherapy-induced alopecia in breast cancer patients. Breast Cancer Res 2013; 15:R81.
  38. van den Hurk CJ, Peerbooms M, van de Poll-Franse LV, et al. Scalp cooling for hair preservation and associated characteristics in 1411 chemotherapy patients - results of the Dutch Scalp Cooling Registry. Acta Oncol 2012; 51:497.
  39. Fehr MK, Welter J, Sell W, et al. Sensor-controlled scalp cooling to prevent chemotherapy-induced alopecia in female cancer patients. Curr Oncol 2016; 23:e576.
  40. Freites-Martinez A, Shapiro J, Goldfarb S, et al. Hair disorders in patients with cancer. J Am Acad Dermatol 2019; 80:1179.
  41. Saggar V, Wu S, Dickler MN, Lacouture ME. Alopecia with endocrine therapies in patients with cancer. Oncologist 2013; 18:1126.
  42. Freites-Martinez A, Shapiro J, Chan D, et al. Endocrine Therapy-Induced Alopecia in Patients With Breast Cancer. JAMA Dermatol 2018; 154:670.
  43. Hortobagyi GN, Stemmer SM, Burris HA, et al. Ribociclib as First-Line Therapy for HR-Positive, Advanced Breast Cancer. N Engl J Med 2016; 375:1738.
  44. Goetz MP, Toi M, Campone M, et al. MONARCH 3: Abemaciclib As Initial Therapy for Advanced Breast Cancer. J Clin Oncol 2017; 35:3638.
  45. Wu CY, Chen GS, Lan CC. Erosive pustular dermatosis of the scalp after gefitinib and radiotherapy for brain metastases secondary to lung cancer. Clin Exp Dermatol 2008; 33:106.
  46. Donovan JC, Ghazarian DM, Shaw JC. Scarring alopecia associated with use of the epidermal growth factor receptor inhibitor gefitinib. Arch Dermatol 2008; 144:1524.
  47. Murillas R, Larcher F, Conti CJ, et al. Expression of a dominant negative mutant of epidermal growth factor receptor in the epidermis of transgenic mice elicits striking alterations in hair follicle development and skin structure. EMBO J 1995; 14:5216.
  48. Escudier B, Eisen T, Stadler WM, et al. Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med 2007; 356:125.
  49. Llovet JM, Ricci S, Mazzaferro V, et al. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med 2008; 359:378.
  50. Ratain MJ, Eisen T, Stadler WM, et al. Phase II placebo-controlled randomized discontinuation trial of sorafenib in patients with metastatic renal cell carcinoma. J Clin Oncol 2006; 24:2505.
  51. Autier J, Escudier B, Wechsler J, et al. Prospective study of the cutaneous adverse effects of sorafenib, a novel multikinase inhibitor. Arch Dermatol 2008; 144:886.
  52. Rini BI, Escudier B, Tomczak P, et al. Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): a randomised phase 3 trial. Lancet 2011; 378:1931.
  53. Zhang L, Zhou Q, Ma L, et al. Meta-analysis of dermatological toxicities associated with sorafenib. Clin Exp Dermatol 2011; 36:344.
  54. Chang AL, Solomon JA, Hainsworth JD, et al. Expanded access study of patients with advanced basal cell carcinoma treated with the Hedgehog pathway inhibitor, vismodegib. J Am Acad Dermatol 2014; 70:60.
  55. Lear JT, Migden MR, Lewis KD, et al. Long-term efficacy and safety of sonidegib in patients with locally advanced and metastatic basal cell carcinoma: 30-month analysis of the randomized phase 2 BOLT study. J Eur Acad Dermatol Venereol 2018; 32:372.
  56. Minami Y, Minami H, Miyamoto T, et al. Phase I study of glasdegib (PF-04449913), an oral smoothened inhibitor, in Japanese patients with select hematologic malignancies. Cancer Sci 2017; 108:1628.
  57. Tallon B, Blanchard E, Goldberg LJ. Permanent chemotherapy-induced alopecia: case report and review of the literature. J Am Acad Dermatol 2010; 63:333.
  58. Fonia A, Cota C, Setterfield JF, et al. Permanent alopecia in patients with breast cancer after taxane chemotherapy and adjuvant hormonal therapy: Clinicopathologic findings in a cohort of 10 patients. J Am Acad Dermatol 2017; 76:948.
  59. Martín M, de la Torre-Montero JC, López-Tarruella S, et al. Persistent major alopecia following adjuvant docetaxel for breast cancer: incidence, characteristics, and prevention with scalp cooling. Breast Cancer Res Treat 2018; 171:627.
  60. Freites-Martinez A, Shapiro J, van den Hurk C, et al. Hair disorders in cancer survivors. J Am Acad Dermatol 2019; 80:1199.
  61. Núñez-Torres R, Martín M, García-Sáenz JÁ, et al. Association Between ABCB1 Genetic Variants and Persistent Chemotherapy-Induced Alopecia in Women With Breast Cancer. JAMA Dermatol 2020; 156:987.
  62. Vendelbo Johansen L. Scalp hypothermia in the prevention of chemotherapy-induced alopecia. Acta Radiol Oncol 1985; 24:113.
  63. Tollenaar RA, Liefers GJ, Repelaer van Driel OJ, van de Velde CJ. Scalp cooling has no place in the prevention of alopecia in adjuvant chemotherapy for breast cancer. Eur J Cancer 1994; 30A:1448.
  64. Symonds RP, McCormick CV, Maxted KJ. Adriamycin alopecia prevented by cold air scalp cooling. Am J Clin Oncol 1986; 9:454.
  65. Komen MMC, van den Hurk CJG, Nortier JWR, et al. Prolonging the duration of post-infusion scalp cooling in the prevention of anthracycline-induced alopecia: a randomised trial in patients with breast cancer treated with adjuvant chemotherapy. Support Care Cancer 2019; 27:1919.
  66. van den Hurk CJ, Breed WP, Nortier JW. Short post-infusion scalp cooling time in the prevention of docetaxel-induced alopecia. Support Care Cancer 2012; 20:3255.
  67. Komen MM, Breed WP, Smorenburg CH, et al. Results of 20- versus 45-min post-infusion scalp cooling time in the prevention of docetaxel-induced alopecia. Support Care Cancer 2016; 24:2735.
  68. Lugtenberg RT, van den Hurk CJG, Smorenburg CH, et al. Comparable effectiveness of 45- and 20-min post-infusion scalp cooling time in preventing paclitaxel-induced alopecia - a randomized controlled trial. Support Care Cancer 2022; 30:6641.
  69. Komen MMC, Smorenburg CH, Nortier JWR, et al. Results of scalp cooling during anthracycline containing chemotherapy depend on scalp skin temperature. Breast 2016; 30:105.
  70. NCCN Clinical Practice Guidelines in Oncology. Available at: https://www.nccn.org/professionals/physician_gls/default.aspx#site (Accessed on November 04, 2019).
  71. Lacouture ME, Sibaud V, Gerber PA, et al. Prevention and management of dermatological toxicities related to anticancer agents: ESMO Clinical Practice Guidelines☆. Ann Oncol 2021; 32:157.
  72. Komen MM, Smorenburg CH, van den Hurk CJ, Nortier JW. Factors influencing the effectiveness of scalp cooling in the prevention of chemotherapy-induced alopecia. Oncologist 2013; 18:885.
  73. Shin H, Jo SJ, Kim DH, et al. Efficacy of interventions for prevention of chemotherapy-induced alopecia: a systematic review and meta-analysis. Int J Cancer 2015; 136:E442.
  74. Parker R. The effectiveness of scalp hypothermia in preventing cyclophosphamide-induced alopecia. Oncol Nurs Forum 1987; 14:49.
  75. Knobf M, Kalm D, Mealia M. Clinical observations of scalp cooling in patients receiving multidrug chemotherapy. Oncol Nurs Forum 1989; 16(suppl):200.
  76. Middleton J, Franks D, Buchanan RB, et al. Failure of scalp hypothermia to prevent hair loss when cyclophosphamide is added to doxorubicin and vincristine. Cancer Treat Rep 1985; 69:373.
  77. Wheelock JB, Myers MB, Krebs HB, Goplerud DR. Ineffectiveness of scalp hypothermia in the prevention of alopecia in patients treated with doxorubicin and cisplatin combinations. Cancer Treat Rep 1984; 68:1387.
  78. Wang S, Yang T, Shen A, et al. 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.
  79. Rugo HS, Klein P, Melin SA, et al. Association Between Use of a Scalp Cooling Device and Alopecia After Chemotherapy for Breast Cancer. JAMA 2017; 317:606.
  80. Nangia J, Wang T, Osborne C, et al. Effect of a Scalp Cooling Device on Alopecia in Women Undergoing Chemotherapy for Breast Cancer: The SCALP Randomized Clinical Trial. JAMA 2017; 317:596.
  81. Munzone E, Bagnardi V, Campennì G, et al. Preventing chemotherapy-induced alopecia: a prospective clinical trial on the efficacy and safety of a scalp-cooling system in early breast cancer patients treated with anthracyclines. Br J Cancer 2019; 121:325.
  82. Nangia J. Quality of Life Matters: It Is Time to Integrate Scalp Cooling in Routine Clinical Practice. J Oncol Pract 2018; 14:157.
  83. Lemenager M, Lecomte S, Bonneterre ME, et al. Effectiveness of cold cap in the prevention of docetaxel-induced alopecia. Eur J Cancer 1997; 33:297.
  84. Grevelman EG, Breed WP. Prevention of chemotherapy-induced hair loss by scalp cooling. Ann Oncol 2005; 16:352.
  85. Macduff C, Mackenzie T, Hutcheon A, et al. The effectiveness of scalp cooling in preventing alopecia for patients receiving epirubicin and docetaxel. Eur J Cancer Care (Engl) 2003; 12:154.
  86. Massey CS. A multicentre study to determine the efficacy and patient acceptability of the Paxman Scalp Cooler to prevent hair loss in patients receiving chemotherapy. Eur J Oncol Nurs 2004; 8:121.
  87. Cigler T, Isseroff D, Fiederlein B, et al. Efficacy of Scalp Cooling in Preventing Chemotherapy-Induced Alopecia in Breast Cancer Patients Receiving Adjuvant Docetaxel and Cyclophosphamide Chemotherapy. Clin Breast Cancer 2015; 15:332.
  88. Belum VR, de Barros Silva G, Laloni MT, et al. Cold thermal injury from cold caps used for the prevention of chemotherapy-induced alopecia. Breast Cancer Res Treat 2016; 157:395.
  89. Breed WP. Response to "Hair 'regrowth' during chemotherapy after scalp cooling technique". Int J Dermatol 2016; 55:e465.
  90. Schaffrin-Nabe D, Schmitz I, Josten-Nabe A, et al. The Influence of Various Parameters on the Success of Sensor-Controlled Scalp Cooling in Preventing Chemotherapy-Induced Alopecia. Oncol Res Treat 2015; 38:489.
  91. van den Hurk CJ, van den Akker-van Marle ME, Breed WP, et al. Impact of scalp cooling on chemotherapy-induced alopecia, wig use and hair growth of patients with cancer. Eur J Oncol Nurs 2013; 17:536.
  92. Sahadevan SW, Ding SR, Del Priore G. Hair "regrowth" during chemotherapy after scalp cooling technique. Int J Dermatol 2016; 55:e463.
  93. Fushimi A, Shinozaki N, Takeyama H. Hair regrowth using a properly fitted scalp cooling cap during adjuvant chemotherapy for breast cancer. Int Cancer Conf J 2019; 8:181.
  94. Satterwhite B, Zimm S. The use of scalp hypothermia in the prevention of doxorubicin-induced hair loss. Cancer 1984; 54:34.
  95. Giaccone G, Di Giulio F, Morandini MP, Calciati A. Scalp hypothermia in the prevention of doxorubicin-induced hair loss. Cancer Nurs 1988; 11:170.
  96. Witman G, Cadman E, Chen M. Misuse of scalp hypothermia. Cancer Treat Rep 1981; 65:507.
  97. Rugo HS, Melin SA, Voigt J. Scalp cooling with adjuvant/neoadjuvant chemotherapy for breast cancer and the risk of scalp metastases: systematic review and meta-analysis. Breast Cancer Res Treat 2017; 163:199.
  98. Forsberg SA. Scalp cooling therapy and cytotoxic treatment. Lancet 2001; 357:1134.
  99. van den Hurk CJ, van de Poll-Franse LV, Breed WP, et al. Scalp cooling to prevent alopecia after chemotherapy can be considered safe in patients with breast cancer. Breast 2013; 22:1001.
  100. Lemieux J, Amireault C, Provencher L, Maunsell E. Incidence of scalp metastases in breast cancer: a retrospective cohort study in women who were offered scalp cooling. Breast Cancer Res Treat 2009; 118:547.
  101. Who should NOT use The DigniCap Scalp Cooling System? (What are the contraindications?). Available at: https://support.dignicap.com/support/solutions/articles/9000134677-who-should-not-use-the-dignicap-scalp-cooling-system-what-are-the-contraindications- (Accessed on October 28, 2019).
  102. van den Hurk C, de Beer F, Dries W, et al. No prevention of radiotherapy-induced alopecia by scalp cooling. Radiother Oncol 2015; 117:193.
  103. Novice M, Novice T, Henry NL, et al. Identifying Barriers and Facilitators to Scalp Cooling Therapy Through a National Survey of the Awareness, Practice Patterns, and Attitudes of Oncologists. JCO Oncol Pract 2022; 18:e225.
  104. Hershman DL. Scalp Cooling to Prevent Chemotherapy-Induced Alopecia: The Time Has Come. JAMA 2017; 317:587.
  105. West HJ. Do the Data on Scalp Cooling for Patients With Breast Cancer Warrant Broad Adoption? JAMA Oncol 2017.
  106. Katz A. Scalp Cooling: The Prevention of Chemotherapy-Induced Alopecia
. Clin J Oncol Nurs 2017; 21:413.
  107. Janssen FE, Van Leeuwen GM, Van Steenhoven AA. Modelling of temperature and perfusion during scalp cooling. Phys Med Biol 2005; 50:4065.
  108. Araoye EF, Stearns V, Aguh C. Considerations for the Use of Scalp Cooling Devices in Black Patients. J Clin Oncol 2020; 38:3575.
  109. Dilawari A, Gallagher C, Alintah P, et al. Does Scalp Cooling Have the Same Efficacy in Black Patients Receiving Chemotherapy for Breast Cancer? Oncologist 2021; 26:292.
  110. Fundamentals of Ethnic Hair: The Dermatologists' Perspective, Aguh C, Okoye GA (Eds), Springer International, Cham, Switzerland 2017.
  111. Rodriguez R, Machiavelli M, Leone B, et al. Minoxidil (Mx) as a prophylaxis of doxorubicin--induced alopecia. Ann Oncol 1994; 5:769.
  112. Duvic M, Lemak NA, Valero V, et al. A randomized trial of minoxidil in chemotherapy-induced alopecia. J Am Acad Dermatol 1996; 35:74.
  113. Kuo AM, et al. Oral minoxidil for the treatment of late alopecia in cancer survivors (abstract). J Clin Oncol 40, 2022 (suppl 16; abstr 12022). Abstract available online at https://meetings.asco.org/abstracts-presentations/209866 (Accessed on August 18, 2022).
  114. Kang J, Lee JW, Kwon O. Efficacy of low-dose oral minoxidil in the management of anticancer therapy-induced alopecia in patients with breast cancer: A retrospective cohort study. J Am Acad Dermatol 2023; 88:1170.
  115. Glaser DA, Hossain P, Perkins W, et al. 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.
  116. Rozner RN, Freites-Martinez A, Shapiro J, et al. Safety of 5α-reductase inhibitors and spironolactone in breast cancer patients receiving endocrine therapies. Breast Cancer Res Treat 2019; 174:15.
  117. Bommareddy K, Hamade H, Lopez-Olivo MA, et al. Association of Spironolactone Use With Risk of Cancer: A Systematic Review and Meta-analysis. JAMA Dermatol 2022; 158:275.
  118. Jimenez JJ, Yunis AA. Protection from chemotherapy-induced alopecia by 1,25-dihydroxyvitamin D3. Cancer Res 1992; 52:5123.
  119. Jimenez JJ, Alvarez E, Bustamante CD, Yunis AA. Pretreatment with 1,25(OH)2D3 protects from Cytoxan-induced alopecia without protecting the leukemic cells from Cytoxan. Am J Med Sci 1995; 310:43.
  120. Hidalgo M, Rinaldi D, Medina G, et al. A phase I trial of topical topitriol (calcitriol, 1,25-dihydroxyvitamin D3) to prevent chemotherapy-induced alopecia. Anticancer Drugs 1999; 10:393.
  121. Reichel H, Koeffler HP, Norman AW. The role of the vitamin D endocrine system in health and disease. N Engl J Med 1989; 320:980.
  122. Lodewijckx J, Robijns J, Claes M, et al. The use of photobiomodulation therapy for the management of chemotherapy-induced alopecia: a randomized, controlled trial (HAIRLASER trial). Support Care Cancer 2023; 31:269.
Topic 1156 Version 52.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟