Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Oct 1.
Published in final edited form as: J Am Acad Dermatol. 2020 Apr 29;83(4):1130–1143. doi: 10.1016/j.jaad.2020.04.105

Dermatologic toxicities to immune checkpoint inhibitor therapy: A review of histopathologic features

Samantha Ellis 1, Aren T Vierra 1, Jillian W Millsop 2, Mario E Lacouture 3, Maija Kiuru 4
PMCID: PMC7492441  NIHMSID: NIHMS1588857  PMID: 32360716

Abstract

Antineoplastic agents that utilize the immune system have revolutionized cancer treatment. Specifically, implementation of immune checkpoint inhibitors, monoclonal antibodies that block cytotoxic T lymphocyte-associated antigen-4 (CTLA-4), programmed cell death protein 1 (PD-1), or programmed death ligand 1 (PD-L1), show improved and sustained responses in cancer patients. However, these agents are associated with a plethora of adverse events, many manifesting in the skin. As the clinical application of cancer immunotherapies expands, understanding the clinical and histopathologic features of associated cutaneous toxicities becomes increasingly important to dermatologists, oncologists, and pathologists to ensure timely diagnosis and appropriate care. This review discusses cutaneous reactions to immune checkpoint inhibitors, focusing on histopathologic features.

Keywords: checkpoint inhibitor, immunotherapy, CTLA-4, PD1, PD-L1, ipilimumab, nivolumab, pembrolizumab, atezolizumab, avelumab, durvalumab, cutaneous, toxicity, adverse event, rash, skin, lichenoid dermatitis, bullous pemphigoid

CAPSULE SUMMARY

  • • Immune checkpoint inhibitors have revolutionized cancer treatment, but can lead to a variety of cutaneous toxicities that may influence decisions to continue therapy

  • • Recognizing the various cutaneous reactions to immune checkpoint blockade, as well as their associated histopathologic findings, is imperative for accurate diagnosis and appropriate patient care.

INTRODUCTION

Immune checkpoint blockade has transformed cancer treatment by enabling sustained responses in cancer patients.1 Checkpoint blockade, including monoclonal antibodies that bind cytotoxic T lymphocyte-associated antigen-4 (CTLA-4), programmed cell death 1 (PD-1), or programmed cell death ligand 1 (PD-L1), inhibits the down-regulation of cytotoxic T lymphocytes, shifting the immune system to an activated, anticancer state.1, 2 However, checkpoint inhibition can lead to numerous adverse events (AEs), often manifesting in the skin. As the use of checkpoint inhibitor therapy continues to expand, delineating the clinical and histopathologic findings of various cutaneous toxicities secondary to checkpoint inhibition helps improve early and accurate diagnosis and guide therapeutic interventions.

CHECKPOINT INHIBITORS

Checkpoints maintain immunologic homeostasis by limiting T lymphocyte activity toward host antigens but can also inadvertently decrease immune surveillance of cancer cells.35 CTLA-4, expressed on the cell surface of activated T cells, prevents continued T cell activation when bound to costimulatory signals. Ipilimumab blocks this interaction allowing the immune system to activate against neoplastic cells.69 Similarly, binding of PD-1 expressed on activated T cells prevents T cell proliferation and excessive inflammatory responses, which tumor and stromal cells evade by expressing PD-1 ligands (PD-L1, PD-L2).3, 1015 By deploying PD-1 or PD-L1 inhibitors that block this interaction, T cells remain unsuppressed carrying out antitumor activity.1618 CTLA-4 inhibitor ipilimumab was approved as the first checkpoint inhibitor in 2011 by the US Food and Drug Administration (FDA) for treatment of metastatic melanoma,19, 20 followed by PD-1 inhibitors nivolumab and pembrolizumab in 2014, and PD-L1 inhibitors atezolizumab in 2016, and durvalumab and avelumab in 2017 to treat various solid organ malignancies.21

Checkpoint inhibition can lead to numerous adverse events, often immune-related (immune-related adverse events [IRAEs]), manifesting in the gastrointestinal tract, liver, and skin, although any organ may be affected. Patient characteristics, like cytokine profiles and HLA types, may be predictive of IRAEs, including pruritus, but their specific influence on cutaneous eruptions remains largely unknown.22, 23 Interestingly, patients who develop dermatologic AEs may demonstrate greater therapeutic responses and outcomes.2426 Cutaneous toxicities are prevalent among all checkpoint inhibitor therapies but appear twice as often during anti-CTLA-4 therapy compared with PD-1 and PD-L1 inhibitors, in 60% versus 20% of patients, respectively.1, 2729,18, 3038 Cutaneous toxicities often manifest earlier than other AEs, generally within three to six weeks after starting ipilimumab and five to nine weeks after PD-1 and PD-L1 inhibitors, though may occur months after initiation of therapy.1, 24, 27, 28, 3941 Most cutaneous AEs are low-grade, with fewer than 3% progressing to a grade 3 or 4 reaction (for Common Terminology Criteria for Adverse Events, see Appendix), and even fewer with PD-1 and PD-L1 inhibitors.3335, 39 In general, maculopapular eruptions are reported most commonly, followed by pruritus and vitiligo, though many other reactions can occur as discussed below.35, 4245 Lastly, while these reactions occur after initiation of therapy, a subset of them may be incidental occurrences, paraneoplastic phenomena, or related to the patient’s past personal or family history, introducing a bias, as well as a limitation of this study, that should be taken into consideration when evaluating these patients.

CUTANEOUS TOXICITIES

Inflammatory reactions

Predominantly superficial perivascular dermatitis

Maculopapular eruptions, occurring in up to 60% of patients treated with CTLA-4 inhibitor therapy, typically show superficial perivascular dermatitis on histopathology. Perivascular dermatitis, occasionally with eosinophils, may occur during PD-1 blockade but is less common.46 Patients demonstrate variably pruritic, erythematous macules and dome-shaped papules, some of which coalesce into patches and plaques.42, 44, 45, 47 Reticulated patterns or koebnerization can be seen.43, 44 Eruptions usually present on the trunk and/or extremities, often on extensor surfaces.43, 44, 47 Rarely, flexural skin, scalp, palms, and face are involved.44, 48 Onset varies from 3 days to 3 weeks after treatment initiation.43, 47, 49

Varying densities of superficial perivascular lymphocytes, often associated with interstitial eosinophils, are present (Table 1).4245, 47, 48, 50 Less frequently, concomitant parakeratosis, spongiosis, exocytosis, papillary dermal edema, and deep perivascular lymphocytes can be seen.42, 45, 47, 48, 50 There are increased numbers of CD4+ lymphocytes compared to CD8+ lymphocytes, as well as regulatory T cells.45, 47, 50

Table 1.

Dermatologic toxicities reported with CTLA-4 inhibitor (ipilimumab), PD-1 inhibitor (nivolumab, pembrolizumab) and PD-L1 inhibitor (atezolizumab, avelumab, durvalumab) therapy.

Dermatologic toxicity Histopathologic features Histologic differential diagnosis Quality of evidence (study designs)1
CTLA-4 inhibitor PD1-inhibitor PD-L1 inhibitor
Inflammatory Acantholytic dermatitis ● Acantholysis and dyskeratosis
● Superficial dermal lymphocytic infiltrate, occasionally with interstitial neutrophils and eosinophils
● Predominance of CD4+ T cells
Pemphigus Hailey-Hailey disease Darier disease Acantholytic acanthoma
Acneiform/follic ular dermatitis or rosacea ● For rosacea, perivascular and perifollicular lymphocytes and dilation of superficial blood vessels Acne vulgaris Seborrheic dermatitis Suppurative folliculitis
Acute generalized exanthematous pustulosis ● Collections of subcorneal neutrophils, often with eosinophils Pustular psoriasis Impetigo Candida infection Subcorneal pustular dermatosis ●●
Bullous pemphigoid ● Subepidermal cleft with eosinophils within the blister cavity and dermis
● DIF: Linear C3 or C3 and IgG along the BMZ
● Salt split DIF: Linear C3 or C3 and IgG at the epidermal aspect of the blister
● 11F: often positive on monkey esophagus ELISA: BP180, sometimes BP230
Bullous arthropod reaction Allergic contact dermatitis Drug reaction Pemphigus vulgaris ●● ●●
CD30 lymphomatoid reaction ● CD30-positive lymphocytic infiltrate in dermis Lymphoma Lymphomatoid papulosis
Dermatomyositi s-like reaction Not reported Lupus erythematosus
Drug reaction with eosinophils and systemic symptoms Not reported Histological features are variable, thus differential diagnosis is broad Spongiotic dermatitis Pustular dermatitis Interface dermatitis Interstitial granulomatous dermatitis ●●
Sarcoidal granulomatous dermatitis ● Multifocal nodular collections of epithelioid histiocytes and scant accompanying lymphocytes
● May contain polarizable material
Infection (including tuberculoid leprosy) Foreign body granuloma Sarcoidal variant of granuloma annulare Cutaneous Crohn's disease Necrobiosis lipoidica Granuloma annulare
Interstitial granulomatous dermatitis ● Superficial interstitial dermal histiocytic infiltrate with scant lymphocytes
● No associated epidermal changes, multinucleate d giant cells, mucin deposition or necrobiosis reported
Interstitial granuloma annulare
Lichenoid dermatitis ● Dense band-like dermal lymphocytic infiltrate obscuring the dermal-epidermal junction
● Variable degree of hyperkeratosis, hypergranulos is, dyskeratotic keratinocytes, vacuolar interface alteration, acanthosis, spongiosis and parakeratosis Occasionally inflammation around adnexal structures
● Not uncommonly hyperkeratosi s, wedge-shaped hypergranulos is, dyskeratosis and irregular acanthosis with saw-tooth rete ridges indistinguisha ble from lichen planus
Lichen planus Lichenoid keratosis Lichen nitidus Lichen striatus Fixed drug reaction Discoid lupus erythematosus ●● ●●
Neutrophilic dermatosis of the dorsal hands See Sweet syndrome (below) Infection Vasculitis Pyoderma gangrenosum Granuloma faciale Behceťs disease
Panniculitis ● Septal and lobular inflammatory infiltrates, including lymphocytes, histiocytes, multinucleate d giant cells, rare eosinophils and neutrophils
● Fibrous septal thickening
● Stains for microorganis ms negative
Erythema nodosum Lupus panniculitis Other panniculitides Infection
Photosensitivity ● Spongiosis with eosinophils, parakeratosis and acanthosis Other spongiotic dermatitides
Prurigo nodularis Not reported Verruca vulgaris Pseudocarcino matous hyperplasia Keratoacantho ma
Psoriasis ● Parakeratosis, diminished granular layer, acanthosis, thinning of suprapapillary plates, dilated superficial dermal capillaries, and mononuclear cells in dermis
● Varying degrees of concomitant spongiosis
Chronic spongiotic dermatitis Seborrheic dermatitis Pityriasis rubra pilaris Syphilis Lichen simplex chronicus ●● ●●
Pyoderma gangrenosum ● An ulcer with dermal neutrophilic infiltrates Infection Vasculitis Sweeťs syndrome Granuloma faciale Behceťs disease
Radiation-associated dermatitis Not reported N/A
Sclerodermoid reaction ● Extensive dermal sclerosis with perivascular lymphocytic infiltrates Morphea Sclerodermoid GVHD Chronic porphyria cutanea tarda Keloid Late-stage radiation dermatitis Lichen sclerosus Borrelia infection
Spongiotic dermatitis ● Spongiosis, perivascular inflammatory cell infiltrates Allergic contact dermatitis Atopic dermatitis Psoriasis Stasis dermatitis Id reaction Pityriasis rosea Tinea infection ●●
Stevens-Johnson syndrome/Toxic epidermal necrolysis -like reaction ● Apoptotic keratinocytes and necrosis of the epidermis
● Sparse mononuclear infiltrate in the dermis
● CD8+ T cells within epidermis and at dermal-epidermal junction
● Increased PD-L1 expression on epidermal keratinocytes near T cells
● Upregulation of CXCL9, CXCL10, CXCL11, PRF1, GZMB, and FASLG (anti- PD-1 agents)
● Leukocytoclas tic vasculitis
Erythema multiforme GVHD Lupus erythematosus Dermatomyositis
Superficial perivascular dermatitis ● Superficial perivascular lymphocytic infiltrates with interstitial eosinophils
● Rarely deep dermal lymphocytic perivascular infiltrates, exocytosis, parakeratosis, papillary dermal edema, spongiosis
● Increased numbers of CD4-positive lymphocytes (CTLA-4 inhibitor)
Urticaria Arthropod bite reaction Drug reaction Scabies Urticarial bullous pemphigoid Allergic contact dermatitis Itchy red bump disease ●●
Sweeťs syndrome ● Dense neutrophilic dermal infiltrates, often extending to the subcutis, occasionally with plasma cells and eosinophils
● Prominent papillary dermal edema
● No evidence of infection or leukocytoclast ic vasculitis
Infection Vasculitis Pyoderma gangrenosum Granuloma faciale Behceťs disease
Xerosis ● Not reported Ichthyosis “Invisible” dermatoses (macular amyloidosis, dermal melanocytosis, mastocytosis, anetoderma, vitiligo, tinea infection)
Alopecia Alopecia, non-scarring ● Peribulbar lymphocytic infiltrate
● Predominantly CD4+ T-cells
Androgenetic alopecia Telogen effluvium Syphilitic alopecia
Alteration of melanocytes Nevi with halolike reaction ● Melanocytes surrounded by lichenoid lymphohistioc ytic infiltrates
● Commonly of CD8+ T-cells, with few CD4+ and CD45R0+ cells
Melanoma Lymphoma Lichenoid Keratosis
Vitiligo CD8+ T cells expressing CXCR3 and producing elevated levels of interferon-γ and tumor necrosis factor-α Post-inflammatory hypopigmentation “Invisible” dermatoses (see above) ●●● ●●
Alteration of keratinocytes, including tumors Actinic keratosis Not reported Squamous cell carcinoma in situ
Basal cell carcinoma Not reported Squamous cell carcinoma Sebaceous carcinoma Other adnexal neoplasms
Keratoacanthoma ● Crateriform keratinocytic proliferation
● Squamous cells with glassy-appearing cytoplasm with minimal cytologic atypia
● An associated lichenoid infiltrate composed of CD3+ T cells with scattered CD20+ B cells
Squamous cell carcinoma Pseudocarcino matous hyperplasia Verruca vulgaris Prurigo nodularis
Seborrheic keratosis Not reported Verruca vulgaris Hidroacantho ma simplex Squamous cell carcinoma in situ
Squamous cell carcinoma Not reported Hyperplastic actinic keratosis Keratoacantho ma Pseudocarcino matous hyperplasia
1

Unknown/no reported studies ○; Case report(s) and/or case series ●; Observational studies (one or more case-control, cross sectional, and/or cohort study) ●●; Comprehensive studies (one or more non-randomized controlled trial, randomized control trial, meta-analysis, and/or systematic review) ●●● Direct immunofluorescence (DIF), Complement component 3 (C3), Immunoglobulin G (IgG), Basement membrane zone (BMZ), Indirect immunofluorescence (IIF), Enzyme-linked immunosorbent assay (ELISA), Graft-versus-host disease (GVHD)

Interface dermatitis (vacuolar and/or lichenoid)

Lichenoid dermatitis.

Lichenoid dermatitis is an AE associated with anti-PD-1 and anti-PD-L1 use, and rarely occurs during ipilimumab treatment.28, 29, 35, 38, 45, 5158 Onset is on average 12 weeks after medication initiation, ranging from 1 to 266 days.28 While pruritus is common, the clinical presentation is otherwise broad, ranging from classic lichen planus with flat-topped violaceous papules to a morbilliform eruption,29, 51, 54, 55, 57 and rarely, pustules.57 Trunk and extremities are typically affected, and less commonly palms, soles, and genitalia.56, 57, 59 Oral mucosa may also be involved.29

A band-like lymphohistiocytic infiltrate along the dermal-epidermal junction is present in all checkpoint-inhibitor-associated lichenoid dermatoses, with variable parakeratosis, hypergranulosis, acanthosis, spongiosis, vacuolar interface alteration, dyskeratosis, dermal eosinophils, and melanophages (Figure 1).35, 5357 Subepidermal edema or clefting can occur. Reactions indistinguishable from lichen planus, with wedge-shaped hypergranulosis and saw-tooth rete ridges, are not uncommon.53, 57 Compared to classic lichen planus, histiocyte counts are typically higher with anti-PD-1 therapy, as well as the degree of spongiosis and epidermal necrosis.55 Contrary to mixed CD4+ and CD8+ infiltrates often with predominance of CD8+ infiltrates typically seen in lichen planus, those induced by anti-PD-1 therapy are CD4+ T cell predominant.54, 55

Figure 1.

Figure 1.

Bullous lichenoid dermatitis secondary to nivolumab. Biopsy shows a band-like lymphocytic infiltrate associated with a cleft formation at the dermal-epidermal junction.

Additionally, CD163+ histiocytes are more abundant in immunotherapy-associated reactions, while the percentages of CD3, CD20, PD-1, CD25, Foxp3, CXCL13, and PD-L1 are similar to lichen planus.55 While the epithelial antigen driving the lichenoid response remains unknown, PD-1 inhibitors likely unmask autoreactive T-cells.60, 61 Finally, other dermatoses with a lichenoid infiltrate, including lichen sclerosus, pityriasis lichenoides chronica, and lichen planus pemphigoides have also been reported.55, 59, 62

Stevens-Johnson syndrome (SJS)/Toxic epidermal necrolysis (TEN)-like reaction.

SJS/TEN-like reactions with CTLA-4, PD-1, or PD-L1 inhibitors are rare, but portend a poor prognosis.52, 53, 6369 Patients may present with a morbilliform eruption, eventually developing targetoid patches, epidermal detachment, and mucous membrane ulcerations.64, 65 Importantly, SJS/TEN can have a delayed onset, as most incidents manifest weeks to months after treatment initiation.53, 64, 65, 70

Variable epidermal necrosis is present, associated with vacuolar interface alteration, cleavage along the dermal-epidermal plane, and subepidermal lymphocytes.52, 53, 6366, 70 Leukocytoclastic vasculitis (LCV) has been reported.67, 68 CD8+ T cells are present, as well as increases in PD-L1 expression of lymphocytes and keratinocytes in the epidermis.52, 63 Increased PD-L1 expression may indicate an attempt to counter lymphocyte hyperactivity induced by anti-PD-1 agents.63 Skin toxicities associated with anti-PD-1 agents that show necrotic keratinocytes display characteristic gene expression profiles that resemble SJS/TEN, with upregulation of CXCL9, CXCL10, CXCL11, PRF1, GZMB, and FASLG.52, 63

Psoriasis

Psoriasis is a well-established AE secondary to PD-1 and PD-L1 blockade. It develops days to months after therapy initiation and presents as well-demarcated, scaly, erythematous papules and plaques on trunk and extremities.46, 7176 Guttate, inverse, and palmoplantar presentations have been reported.46, 59, 72, 7577 Individuals with established psoriasis may flare while undergoing treatment.71, 76, 7880

Several classic features of psoriasis are present, including parakeratosis, neutrophils within or beneath stratum corneum, granular layer absence, acanthosis, suprapapillary plate thinning, dilated superficial dermal capillaries, and mononuclear cells in the dermis.46, 59, 7274, 77 Concomitant spongiosis may be seen, especially with inverse presentation similar to classis psoriasis.46, 59, 75 PD-1 blockade appears to cause a shift to a pro-inflammatory Th-1/Th-17 response, increasing levels of interferon-gamma, tumor necrosis factor-alpha (TNF-alpha), and interleukins 2, 6, and 17.81 These changes may contribute to psoriasis in patients undergoing PD-1 inhibitor therapy.72, 73

Acantholytic dermatitis

Acantholytic dermatitis has been reported with CTLA-4 or PD-1 inhibitor therapy or combination therapy.35, 45, 8285 It presents as intensely pruritic erythematous papules or papulovesicles on the trunk and occasionally the proximal extremities.45, 8284 Occasionally, hyperkeratotic, annular or targetoid papules or plaques are present.85

Acantholysis is characteristic, and some cases are accompanied by dyskeratosis resembling Grover’s disease.45, 8284, 86 Dermal lymphocytic infiltrates, occasionally with eosinophils and neutrophils, are present.45, 83, 84 Infiltrates are often band-like when associated with PD-1 inhibitors. Predominance of CD4+ T cells over CD8+ T cells may be noted.83 Direct immunofluorescence (DIF) is typically negative, though one reported case of a paraneoplastic pemphigus-like reaction exists.85 However, acantholytic dermatitis has not been associated with identifiable immunoreactant deposition, circulating autoantibodies, or clinical blistering.

Granulomatous dermatitis

Interstitial granulomatous dermatitis.

Interstitial granulomatous dermatitis is rarely seen secondary to CTLA-4 or PD-1 inhibitor therapy or combination therapy and may be secondary to the cancer itself.45, 87 Interstitial granulomatous dermatitis may present as asymptomatic erythematous papules and plaques on the trunk and extremities shortly after initiating treatment.

Interstitial histiocytic infiltrates in the superficial dermis with scant lymphocytes are characteristic. Eosinophils and giant cells may be present. Epidermal changes, mucin deposition, or necrobiosis are absent.45

Sarcoidal granulomatous dermatitis.

Sarcoid-like lesions involving the skin, lungs, and hilar/mediastinal lymph nodes may occur in patients undergoing CTLA-4 or PD-1 inhibitor therapy.8892 Onset is typically at least one month after treatment initiation.91, 9395 As lesions may be clinically or radiographically concerning for cancer recurrence, accurate diagnosis is imperative.29, 92Cutaneous presentation varies from solitary to multiple erythematous to brown papules, plaques, or nodules on the trunk, extremities, or head and neck.8891 Prior scars may be involved.91, 94, 96

Multifocal discrete nodular collections of epithelioid histiocytes with scant accompanying lymphocytes, i.e. sarcoidal granulomas, are present in the dermis, in some cases extending into the subcutis.8890, 94, 95 Polarizable material may be present.89, 97 Infection should be excluded.9395

Acute generalized exanthematous pustulosis (AGEP)

AGEP, occasionally observed with checkpoint inhibitor therapy, presents as diffuse edematous erythema with sterile pustules involving the extremities, trunk, and groin. Collections of subcorneal neutrophils and often eosinophils are characteristic.53, 98

Panniculitis

Panniculitis with clinical erythema nodosum-like features rarely occurs in combination therapy with ipilimumab and nivolumab. It presents as tender nodules on lower extremities and possibly forearms.57

Eruptions show a septal and lobular panniculitis, with fibrous septal thickening and a mixture of lymphocytes, histiocytes, multinucleated giant cells, and rare eosinophils and neutrophils.57 Findings are indistinguishable from erythema nodosum, especially early forms, secondary to other causes. Stains for microorganisms are negative.

Neutrophilic dermatoses

Sweet syndrome.

Sweet syndrome may present during CTLA-4 inhibitor therapy as painful, erythematous and edematous or pseudovesicular papules and plaques.99101 Hands may be exclusively involved (neutrophilic dermatosis of the dorsal hands).101

Papillary dermal edema and dense neutrophilic dermal infiltrates, often extending to the subcutis, are present, without evidence of infection or LCV.99101 Plasma cells, which are a unique finding and may be a distinguishing factor of ipilimumab-induced Sweet syndrome, and eosinophils may be present.99

Pyoderma gangrenosum (PG).

PG is infrequently reported in association with anti-CTLA-4 treatment.48, 102 PG presents as ulceration(s) with violaceous, undermined borders.

Ulceration with dermal neutrophilic infiltrates is characteristic.102 Ipilimumab may cause PG through triggering TNF-alpha from activated NK cells, in addition to lowering regulatory T cell function.103

Immunobullous reactions

Bullous pemphigoid (BP)

BP is another well-established AE associated with PD-1 and PD-L1 inhibition.46, 53, 58, 70, 104110 Onset varies from weeks to several months after therapy initiation.46, 58, 105, 106, 109, 110 Bullous eruptions are often preceded by pruritus and may initially present as non-specific maculopapular or urticarial eruptions.58, 105, 106, 111 Eventually tense bullae and vesicles develop on the trunk and extremities.46, 58, 104111 Mucosal involvement is not uncommon.58, 70, 105, 109

Subepidermal clefting with eosinophils is characteristic, though clefting is not always present (Figure 2). Superficial dermal infiltrates composed of lymphocytes and eosinophils, and occasionally neutrophils are present.58, 70, 104, 105, 111 As with classic BP, DIF demonstrates linear deposits of complement component 3 (C3) and immunoglobulin G (IgG) along the basement membrane zone, localizing to the epidermal aspect of the blister on salt-split DIF.58, 70, 105107 Indirect immunofluorescence (IIF) on monkey esophagus is positive in many cases.58, 105 Enzyme-linked immunosorbent assay (ELISA) detects antibodies against the hemidesmosomal protein BP180, and sometimes BP230 antibodies.58, 105109, 111

Figure 2.

Figure 2.

Bullous pemphigoid secondary to pembrolizumab. Biopsy shows perivascular eosinophils and vacuolar alteration along the junction. Bullae were not present histologically in the biopsy specimen. DIF showed deposition of C3 and IgG along the junction (not shown). Clinically, the patient had intact and eroded bullae on erythematous base.

BP may develop secondary to recognition of common antigens BP180 and BP230 shared between the cutaneous basement membrane and tumor cells.105, 112 Antibody-secreting B cells may also play a role, as PD-1 inhibition can activate B cells and inhibit immunosuppressive B regulatory cells.113 PD-1 blockade may also unmask incipient BP, BP does not resolve in some patients after cessation of checkpoint inhibition.106

Alopecia and other hair abnormalities

Non-scarring alopecia can occur during CTLA-4 or PD-1 inhibitor treatment.44, 49 Non-scarring alopecia associated with ipilimumab may show features of alopecia areata (AA) and be accompanied by signs of autoimmune dysregulation, including hypophysitis and widespread vitiligo.49 A peribulbar, predominantly CD4+ T cell infiltrate with scant CD8+ cells, is present.44 Interestingly, CTLA-4 gene variants are linked with AA.114, 115 In AA mouse models, supplementation with CTLA-4 IgG prevents development of AA.116 In melanoma patients, activated T cells may be targeting melanocyte antigens in the hair bulb, leading to hair loss.117

Repigmentation of gray hair during anti-PD-1 and anti-PD-L1 therapy for non-small cell lung cancer has been observed.118

Alteration of melanocytes

Vitiligo

Vitiligo has the highest level of evidence for association with all checkpoint inhibitor therapy, particularly ipilimumab, occurring in up to 11% of patients with metastatic melanoma.18, 38, 53, 59, 119, 120 Development of vitiligo may be associated with improved treatment response and survival.26, 121 It typically presents with depigmented macules occurring on photoexposed sites and without personal or family history of vitiligo or other autoimmune disorders. Albeit rarely biopsied, the presence of CD8-positive T cells expressing CXCR3 and producing elevated levels of interferon gamma and tumor necrosis factor-alpha has been reported.122 PD-1 inhibitor-associated vitiligo may result from allowing immune effector cells to target a shared antigen among melanoma cells and healthy melanocytes.120, 123

Regression of melanocytic nevi

In addition to tumoral melanosis, i.e. nodular aggregates of melanophages without melanocytes consistent with regression of melanoma,124 regression of melanocytic nevi can happen with anti-CTLA-4 or anti-PD-1 treatments.125, 131

Melanocytes are obscured by lichenoid lymphohistiocytic infiltrates, commonly of CD8+ T cells, with few CD4+ and CD45R0+ cells.125 Melanocytic nevi may express melanoma-related antigens and become targets of anti-CTLA therapy, leading to local destruction by activated T cells.126

Alteration of keratinocytes

Benign, precancerous, and cancerous keratinocytic lesions are rarely associated with PD-1 inhibition.53, 127, 128 These include seborrheic keratosis, actinic keratosis, keratoacanthomas, and squamous cell carcinoma.

Other dermatologic toxicities

Folliculitis, acneiform reactions, or rosacea can occur during CTLA-4 or PD-1 inhibitor therapy.54,6,129,84 Rosacea may present with erythema, papules, and pustules that respond to topical metronidazole and doxycycline.129 Histopathologic features include perivascular and perifollicular lymphocytes, and dilation of superficial blood vessels.129

Sclerodermoid reactions are a rare complication of pembrolizumab therapy, presenting with generalized skin thickening and stiffness and progressive decline in joint flexibility. Histopathologic examination shows extensive dermal sclerosis with perivascular lymphocytes.130

Radiation-associated dermatitis is rarely seen with CTLA-4 or PD-1 inhibitors.130 Other rare cutaneous toxicities related to ipilimumab include dermatomyositis and drug reaction with eosinophils and systemic symptoms (DRESS) and photosensitivity reactions related to PD-1 inhibitors.35, 48, 53, 59, 66, 131134

CONCLUSIONS

Immune checkpoint blockade has demonstrated remarkable outcomes for patients with various types of cancer. Checkpoint inhibitors are associated with a range of cutaneous side effects, highlighting the complexity of the immune response and the importance of clinical-histopathologic correlation in accurate recognition of AEs, allowing for appropriate intervention and patient care.

Funding sources:

Research reported in this publication was supported in part by Dermatology Foundation, through Dermatopathology Career Development Award (MK); National Institute of Arthritis and Musculoskeletal and Skin Diseases of the National Institutes of Health under award number U01AR077511 (Dr. Lacouture); Berg, Lutris, Paxman, Novocure, US Biotest, and Veloce (MEL); and the NIH/NCI Cancer Center Support Grant P30 CA008748 (MEL)

Appendix:

Adverse event: Rash/desquamation; Grade 1: Macular or papular eruption or erythema without associated symptoms; Grade 2: Macular or papular eruption or erythema with pruritus or other associated symptoms or localized desquamation or other lesions covering<50% of body surface area; Grade 3: Severe, generalized erythroderma or macular, papular or vesicular eruption or desquamation covering ≥50% body surface area; Grade 4 Generalized exfoliative, ulcerative, or bullous dermatitis; Grade 5 Death)

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Disclosures: Authors SE, ATV, JM, MK have no conflicts of interest to report. MEL has a consultant/speaking role with Legacy Healthcare Services, Adgero, Amryt, Celldex, Debiopharm, Galderma, Johnson and Johnson, Novocure, Lindi, Merck Sharp and Dohme, BMS, Helsinn, Janssen, Menlo, Novartis, F. Hoffmann-La Roche AG, AbbVie Inc, Boehringer Ingelheim, Allergan, Amgen, E.R. Squibb & Sons LLC, EMD Serono, Astrazeneca, Genentech, Leo, Seattle Genetics, Bayer, Lutris, Pierre Fabre, Paxman Coolers, Adjucare, Dignitana, Biotechspert, Teva, Parexel, OnQuality, Novartis, Harborside, Wiley, Azitra, NCODA and Takeda Millenium.

REFERENCES

  • 1.Postow MA, Callahan MK, Wolchok JD. Immune Checkpoint Blockade in Cancer Therapy. J Clin Oncol 2015;33:1974–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012;12:252–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Zou W, Chen L. Inhibitory B7-family molecules in the tumour microenvironment. Nature reviews Immunology 2008;8:467–77. [DOI] [PubMed] [Google Scholar]
  • 4.Curiel TJ, Coukos G, Zou L, Alvarez X, Cheng P, Mottram P et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med 2004;10:942–9. [DOI] [PubMed] [Google Scholar]
  • 5.Kono K, Kawaida H, Takahashi A, Sugai H, Mimura K, Miyagawa N et al. CD4(+)CD25high regulatory T cells increase with tumor stage in patients with gastric and esophageal cancers. Cancer Immunol Immunother 2006;55:1064–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Chambers CA, Kuhns MS, Egen JG, Allison JP. CTLA-4-mediated inhibition in regulation of T cell responses: mechanisms and manipulation in tumor immunotherapy. Annual review of immunology 2001;19:565–94. [DOI] [PubMed] [Google Scholar]
  • 7.Melero I, Hervas-Stubbs S, Glennie M, Pardoll DM, Chen L. Immunostimulatory monoclonal antibodies for cancer therapy. Nat Rev Cancer 2007;7:95–106. [DOI] [PubMed] [Google Scholar]
  • 8.Wolchok JD, Hodi FS, Weber JS, Allison JP, Urba WJ, Robert C et al. Development of ipilimumab: a novel immunotherapeutic approach for the treatment of advanced melanoma. Annals of the New York Academy of Sciences 2013;1291:1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Morse MA. Technology evaluation: ipilimumab, Medarex/Bristol-Myers Squibb. Curr Opin Mol Ther 2005;7:588–97. [PubMed] [Google Scholar]
  • 10.Ishida Y, Agata Y, Shibahara K, Honjo T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. Embo j 1992;11:3887–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Barber DL, Wherry EJ, Masopust D, Zhu B, Allison JP, Sharpe AH et al. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 2006;439:682–7. [DOI] [PubMed] [Google Scholar]
  • 12.Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annual review of immunology 2008;26:677–704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Callahan MK, Wolchok JD. At the bedside: CTLA-4- and PD-1-blocking antibodies in cancer immunotherapy. Journal of leukocyte biology 2013;94:41–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB et al. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med 2002;8:793–800. [DOI] [PubMed] [Google Scholar]
  • 15.Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med 2000;192:1027–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Topalian SL, Drake CG, Pardoll DM. Targeting the PD-1/B7-H1(PD-L1) pathway to activate anti-tumor immunity. Current opinion in immunology 2012;24:207–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T, Minato N. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PDL1 blockade. Proc Natl Acad Sci U S A 2002;99:12293–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 2012;366:2455–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Lipson EJ, Drake CG. Ipilimumab: an anti-CTLA-4 antibody for metastatic melanoma. Clin Cancer Res 2011;17:6958–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Eggermont AM, Maio M, Robert C. Immune checkpoint inhibitors in melanoma provide the cornerstones for curative therapies. Semin Oncol 2015;42:429–35. [DOI] [PubMed] [Google Scholar]
  • 21.Gong J, Chehrazi-Raffle A, Reddi S, Salgia R. Development of PD-1 and PD-L1 inhibitors as a form of cancer immunotherapy: a comprehensive review of registration trials and future considerations. Journal for Immunotherapy of Cancer 2018;6:8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Lim SY, Lee JH, Gide TN, Menzies AM, Guminski A, Carlino MS et al. Circulating Cytokines Predict Immune-Related Toxicity in Melanoma Patients Receiving Anti-PD-1-Based Immunotherapy. Clin Cancer Res 2019;25:1557–63. [DOI] [PubMed] [Google Scholar]
  • 23.Hasan Ali O, Berner F, Bomze D, Fassler M, Diem S, Cozzio A et al. Human leukocyte antigen variation is associated with adverse events of checkpoint inhibitors. European journal of cancer (Oxford, England : 1990) 2019;107:8–14. [DOI] [PubMed] [Google Scholar]
  • 24.Freeman-Keller M, Kim Y, Cronin H, Richards A, Gibney G, Weber JS. Nivolumab in Resected and Unresectable Metastatic Melanoma: Characteristics of Immune-Related Adverse Events and Association with Outcomes. Clin Cancer Res 2016;22:886–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Sanlorenzo M, Vujic I, Daud A, Algazi A, Gubens M, Luna SA et al. Pembrolizumab Cutaneous Adverse Events and Their Association With Disease Progression. JAMA Dermatol 2015;151:1206–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Teulings HE, Limpens J, Jansen SN, Zwinderman AH, Reitsma JB, Spuls PI et al. Vitiligo-like depigmentation in patients with stage III-IV melanoma receiving immunotherapy and its association with survival: a systematic review and meta-analysis. J Clin Oncol 2015;33:773–81. [DOI] [PubMed] [Google Scholar]
  • 27.Naidoo J, Page DB, Li BT, Connell LC, Schindler K, Lacouture ME et al. Toxicities of the anti-PD-1 and anti-PD-L1 immune checkpoint antibodies. Ann Oncol 2015;26:2375–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Curry JL, Tetzlaff MT, Nagarajan P, Drucker C, Diab A, Hymes SR et al. Diverse types of dermatologic toxicities from immune checkpoint blockade therapy. J Cutan Pathol 2017;44:158–76. [DOI] [PubMed] [Google Scholar]
  • 29.Sibaud V Dermatologic Reactions to Immune Checkpoint Inhibitors : Skin Toxicities and Immunotherapy. Am J Clin Dermatol 2018;19:345–61. [DOI] [PubMed] [Google Scholar]
  • 30.Villadolid J, Amin A. Immune checkpoint inhibitors in clinical practice: update on management of immune-related toxicities. Translational lung cancer research 2015;4:560–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.O’Day SJ, Maio M, Chiarion-Sileni V, Gajewski TF, Pehamberger H, Bondarenko IN et al. Efficacy and safety of ipilimumab monotherapy in patients with pretreated advanced melanoma: a multicenter single-arm phase II study. Ann Oncol 2010;21:1712–7. [DOI] [PubMed] [Google Scholar]
  • 32.Weber J, Thompson JA, Hamid O, Minor D, Amin A, Ron I et al. A randomized, double-blind, placebo-controlled, phase II study comparing the tolerability and efficacy of ipilimumab administered with or without prophylactic budesonide in patients with unresectable stage III or IV melanoma. Clin Cancer Res 2009;15:5591–8. [DOI] [PubMed] [Google Scholar]
  • 33.Kanz BA, Pollack MH, Johnpulle R, Puzanov I, Horn L, Morgans A et al. Safety and efficacy of anti-PD-1 in patients with baseline cardiac, renal, or hepatic dysfunction. Journal for immunotherapy of cancer 2016;4:60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 2012;366:2443–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Belum VR, Benhuri B, Postow MA, Hellmann MD, Lesokhin AM, Segal NH et al. Characterisation and management of dermatologic adverse events to agents targeting the PD-1 receptor. Eur J Cancer 2016;60:12–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Antonia SJ, Villegas A, Daniel D, Vicente D, Murakami S, Hui R et al. Durvalumab after Chemoradiotherapy in Stage III Non-Small-Cell Lung Cancer. N Engl J Med 2017;377:1919–29. [DOI] [PubMed] [Google Scholar]
  • 37.Rittmeyer A, Barlesi F, Waterkamp D, Park K, Ciardiello F, von Pawel J et al. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. Lancet 2017;389:255–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Sibaud V, Meyer N, Lamant L, Vigarios E, Mazieres J, Delord JP. Dermatologic complications of anti-PD-1/PD-L1 immune checkpoint antibodies. Curr Opin Oncol 2016;28:254–63. [DOI] [PubMed] [Google Scholar]
  • 39.Minkis K, Garden BC, Wu S, Pulitzer MP, Lacouture ME. The risk of rash associated with ipilimumab in patients with cancer: a systematic review of the literature and meta-analysis. J Am Acad Dermatol 2013;69:e121–8. [DOI] [PubMed] [Google Scholar]
  • 40.Weber JS, Hodi FS, Wolchok JD, Topalian SL, Schadendorf D, Larkin J et al. Safety Profile of Nivolumab Monotherapy: A Pooled Analysis of Patients With Advanced Melanoma. J Clin Oncol 2017;35:785–92. [DOI] [PubMed] [Google Scholar]
  • 41.Weber JS, Kahler KC, Hauschild A. Management of immune-related adverse events and kinetics of response with ipilimumab. J Clin Oncol 2012;30:2691–7. [DOI] [PubMed] [Google Scholar]
  • 42.Attia P, Phan GQ, Maker AV, Robinson MR, Quezado MM, Yang JC et al. Autoimmunity correlates with tumor regression in patients with metastatic melanoma treated with anti-cytotoxic T-lymphocyte antigen-4. J Clin Oncol 2005;23:6043–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Hodi FS, Mihm MC, Soiffer RJ, Haluska FG, Butler M, Seiden MV et al. Biologic activity of cytotoxic T lymphocyte-associated antigen 4 antibody blockade in previously vaccinated metastatic melanoma and ovarian carcinoma patients. Proc Natl Acad Sci U S A 2003;100:4712–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Jaber SH, Cowen EW, Haworth LR, Booher SL, Berman DM, Rosenberg SA et al. Skin reactions in a subset of patients with stage IV melanoma treated with anti-cytotoxic T-lymphocyte antigen 4 monoclonal antibody as a single agent. Arch Dermatol 2006;142:166–72. [DOI] [PubMed] [Google Scholar]
  • 45.Perret RE, Josselin N, Knol AC, Khammari A, Cassecuel J, Peuvrel L et al. Histopathological aspects of cutaneous erythematous-papular eruptions induced by immune checkpoint inhibitors for the treatment of metastatic melanoma. International journal of dermatology 2017;56:527–33. [DOI] [PubMed] [Google Scholar]
  • 46.Kaunitz GJ, Loss M, Rizvi H, Ravi S, Cuda JD, Bleich KB et al. Cutaneous Eruptions in Patients Receiving Immune Checkpoint Blockade: Clinicopathologic Analysis of the Nonlichenoid Histologic Pattern. Am J Surg Pathol 2017;41:1381–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Phan GQ, Yang JC, Sherry RM, Hwu P, Topalian SL, Schwartzentruber DJ et al. Cancer regression and autoimmunity induced by cytotoxic T lymphocyte-associated antigen 4 blockade in patients with metastatic melanoma. Proc Natl Acad Sci U S A 2003;100:8372–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Voskens CJ, Goldinger SM, Loquai C, Robert C, Kaehler KC, Berking C et al. The price of tumor control: an analysis of rare side effects of anti-CTLA-4 therapy in metastatic melanoma from the ipilimumab network. PLoS One 2013;8:e53745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Assi H, Wilson KS. Immune toxicities and long remission duration after ipilimumab therapy for metastatic melanoma: two illustrative cases. Current oncology (Toronto, Ont) 2013;20:e165–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Sanderson K, Scotland R, Lee P, Liu D, Groshen S, Snively J et al. Autoimmunity in a phase I trial of a fully human anti-cytotoxic T-lymphocyte antigen-4 monoclonal antibody with multiple melanoma peptides and Montanide ISA 51 for patients with resected stages III and IV melanoma. J Clin Oncol 2005;23:741–50. [DOI] [PubMed] [Google Scholar]
  • 51.Chou S, Hwang SJ, Carlos G, Wakade D, Fernandez-Penas P. Histologic Assessment of Lichenoid Dermatitis Observed in Patients With Advanced Malignancies on Antiprogramed Cell Death-1 (anti-PD-1) Therapy With or Without Ipilimumab. Am J Dermatopathol 2017;39:23–7. [DOI] [PubMed] [Google Scholar]
  • 52.Goldinger SM, Stieger P, Meier B, Micaletto S, Contassot E, French LE et al. Cytotoxic Cutaneous Adverse Drug Reactions during Anti-PD-1 Therapy. Clin Cancer Res 2016;22:4023–9. [DOI] [PubMed] [Google Scholar]
  • 53.Hwang SJ, Carlos G, Wakade D, Byth K, Kong BY, Chou S et al. Cutaneous adverse events (AEs) of anti-programmed cell death (PD)-1 therapy in patients with metastatic melanoma: A single-institution cohort. J Am Acad Dermatol 2016;74:455–61.e1. [DOI] [PubMed] [Google Scholar]
  • 54.Joseph RW, Cappel M, Goedjen B, Gordon M, Kirsch B, Gilstrap C et al. Lichenoid dermatitis in three patients with metastatic melanoma treated with anti-PD-1 therapy. Cancer immunology research 2015;3:18–22. [DOI] [PubMed] [Google Scholar]
  • 55.Schaberg KB, Novoa RA, Wakelee HA, Kim J, Cheung C, Srinivas S et al. Immunohistochemical analysis of lichenoid reactions in patients treated with anti-PD-L1 and anti-PD-1 therapy. J Cutan Pathol 2016;43:339–46. [DOI] [PubMed] [Google Scholar]
  • 56.Shi VJ, Rodic N, Gettinger S, Leventhal JS, Neckman JP, Girardi M et al. Clinical and Histologic Features of Lichenoid Mucocutaneous Eruptions Due to Anti-Programmed Cell Death 1 and Anti-Programmed Cell Death Ligand 1 Immunotherapy. JAMA Dermatol 2016;152:1128–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Tetzlaff MT, Nagarajan P, Chon S, Huen A, Diab A, Omar P et al. Lichenoid Dermatologic Toxicity From Immune Checkpoint Blockade Therapy: A Detailed Examination of the Clinicopathologic Features. Am J Dermatopathol 2017;39:121–9. [DOI] [PubMed] [Google Scholar]
  • 58.Siegel J, Totonchy M, Damsky W, Berk-Krauss J, Castiglione F Jr., Sznol M et al. Bullous disorders associated with anti-PD-1 and anti-PD-L1 therapy: A retrospective analysis evaluating the clinical and histopathologic features, frequency, and impact on cancer therapy. J Am Acad Dermatol 2018. [DOI] [PubMed] [Google Scholar]
  • 59.Hofmann L, Forschner A, Loquai C, Goldinger SM, Zimmer L, Ugurel S et al. Cutaneous, gastrointestinal, hepatic, endocrine, and renal side-effects of anti-PD-1 therapy. Eur J Cancer 2016;60:190–209. [DOI] [PubMed] [Google Scholar]
  • 60.Roopashree MR, Gondhalekar RV, Shashikanth MC, George J, Thippeswamy SH, Shukla A. Pathogenesis of oral lichen planus--a review. Journal of oral pathology & medicine : official publication of the International Association of Oral Pathologists and the American Academy of Oral Pathology 2010;39:729–34. [DOI] [PubMed] [Google Scholar]
  • 61.Okiyama N, Fujimoto M. Clinical perspectives and murine models of lichenoid tissue reaction/interface dermatitis. Journal of dermatological science 2015;78:167–72. [DOI] [PubMed] [Google Scholar]
  • 62.Mutgi KA, Milhem M, Swick BL, Liu V. Pityriasis lichenoides chronica-like drug eruption developing during pembrolizumab treatment for metastatic melanoma. JAAD Case Rep 2016;2:343–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Vivar KL, Deschaine M, Messina J, Divine JM, Rabionet A, Patel N et al. Epidermal programmed cell death-ligand 1 expression in TEN associated with nivolumab therapy. J Cutan Pathol 2017;44:381–4. [DOI] [PubMed] [Google Scholar]
  • 64.Saw S, Lee HY, Ng QS. Pembrolizumab-induced Stevens-Johnson syndrome in nonmelanoma patients. Eur J Cancer 2017;81:237–9. [DOI] [PubMed] [Google Scholar]
  • 65.Nayar N, Briscoe K, Fernandez Penas P. Toxic Epidermal Necrolysis-like Reaction With Severe Satellite Cell Necrosis Associated With Nivolumab in a Patient With Ipilimumab Refractory Metastatic Melanoma. Journal of immunotherapy (Hagerstown, Md : 1997) 2016;39:149–52. [DOI] [PubMed] [Google Scholar]
  • 66.Dika E, Ravaioli GM, Fanti PA, Piraccini BM, Lambertini M, Chessa MA et al. Cutaneous adverse effects during ipilimumab treatment for metastatic melanoma: a prospective study. Eur J Dermatol 2017;27:266–70. [DOI] [PubMed] [Google Scholar]
  • 67.Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 2010;363:711–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Lacouture ME, Wolchok JD, Yosipovitch G, Kahler KC, Busam KJ, Hauschild A. Ipilimumab in patients with cancer and the management of dermatologic adverse events. J Am Acad Dermatol 2014;71:161–9. [DOI] [PubMed] [Google Scholar]
  • 69.Chirasuthat P, Chayavichitsilp P. Atezolizumab-Induced Stevens-Johnson Syndrome in a Patient with Non-Small Cell Lung Carcinoma. Case Rep Dermatol 2018;10:198–202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Jour G, Glitza IC, Ellis RM, Torres-Cabala CA, Tetzlaff MT, Li JY et al. Autoimmune dermatologic toxicities from immune checkpoint blockade with anti-PD-1 antibody therapy: a report on bullous skin eruptions. J Cutan Pathol 2016;43:688–96. [DOI] [PubMed] [Google Scholar]
  • 71.Chia PL, John T. Severe Psoriasis Flare After Anti-Programmed Death Ligand 1 (PDL1) Therapy for Metastatic Non-Small Cell Lung Cancer (NSCLC). Journal of immunotherapy (Hagerstown, Md : 1997) 2016;39:202–4. [DOI] [PubMed] [Google Scholar]
  • 72.Law-Ping-Man S, Martin A, Briens E, Tisseau L, Safa G. Psoriasis and psoriatic arthritis induced by nivolumab in a patient with advanced lung cancer. Rheumatology (Oxford, England) 2016;55:2087–9. [DOI] [PubMed] [Google Scholar]
  • 73.Murata S, Kaneko S, Harada Y, Aoi N, Morita E. Case of de novo psoriasis possibly triggered by nivolumab. J Dermatol 2017;44:99–100. [DOI] [PubMed] [Google Scholar]
  • 74.Ohtsuka M, Miura T, Mori T, Ishikawa M, Yamamoto T. Occurrence of Psoriasiform Eruption During Nivolumab Therapy for Primary Oral Mucosal Melanoma. JAMA Dermatol 2015;151:797–9. [DOI] [PubMed] [Google Scholar]
  • 75.Totonchy MB, Ezaldein HH, Ko CJ, Choi JN. Inverse Psoriasiform Eruption During Pembrolizumab Therapy for Metastatic Melanoma. JAMA Dermatol 2016;152:590–2. [DOI] [PubMed] [Google Scholar]
  • 76.Bonigen J, Raynaud-Donzel C, Hureaux J, Kramkimel N, Blom A, Jeudy G et al. Anti-PD1-induced psoriasis: a study of 21 patients. J Eur Acad Dermatol Venereol 2017;31:e254–e7. [DOI] [PubMed] [Google Scholar]
  • 77.Ruiz-Banobre J, Abdulkader I, Anido U, Leon L, Lopez-Lopez R, Garcia-Gonzalez J. Development of de novo psoriasis during nivolumab therapy for metastatic renal cell carcinoma: immunohistochemical analyses and clinical outcome. APMIS : acta pathologica, microbiologica, et immunologica Scandinavica 2017;125:259–63. [DOI] [PubMed] [Google Scholar]
  • 78.Menzies AM, Johnson DB, Ramanujam S, Atkinson VG, Wong ANM, Park JJ et al. Anti-PD-1 therapy in patients with advanced melanoma and preexisting autoimmune disorders or major toxicity with ipilimumab. Ann Oncol 2017;28:368–76. [DOI] [PubMed] [Google Scholar]
  • 79.Kato Y, Otsuka A, Miyachi Y, Kabashima K. Exacerbation of psoriasis vulgaris during nivolumab for oral mucosal melanoma. J Eur Acad Dermatol Venereol 2016;30:e89–e91. [DOI] [PubMed] [Google Scholar]
  • 80.Matsumura N, Ohtsuka M, Kikuchi N, Yamamoto T. Exacerbation of Psoriasis During Nivolumab Therapy for Metastatic Melanoma. Acta Derm Venereol 2016;96:259–60. [DOI] [PubMed] [Google Scholar]
  • 81.Dulos J, Carven GJ, van Boxtel SJ, Evers S, Driessen-Engels LJ, Hobo W et al. PD-1 blockade augments Th1 and Th17 and suppresses Th2 responses in peripheral blood from patients with prostate and advanced melanoma cancer. Journal of immunotherapy (Hagerstown, Md : 1997) 2012;35:169–78. [DOI] [PubMed] [Google Scholar]
  • 82.Munoz J, Guillot B, Girard C, Dereure O, Du-Thanh A. First report of ipilimumab-induced Grover disease. Br J Dermatol 2014;171:1236–7. [DOI] [PubMed] [Google Scholar]
  • 83.Uemura M, Fa’ak F, Haymaker C, McQuail N, Sirmans E, Hudgens CW et al. A case report of Grover’s disease from immunotherapy-a skin toxicity induced by inhibition of CTLA-4 but not PD-1. Journal for immunotherapy of cancer 2016;4:55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Koelzer VH, Buser T, Willi N, Rothschild SI, Wicki A, Schiller P et al. Grover’s-like drug eruption in a patient with metastatic melanoma under ipilimumab therapy. Journal for immunotherapy of cancer 2016;4:47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Chen WS, Tetzlaff MT, Diwan H, Jahan-Tigh R, Diab A, Nelson K et al. Suprabasal acantholytic dermatologic toxicities associated checkpoint inhibitor therapy: A spectrum of immune reactions from paraneoplastic pemphigus-like to Grover-like lesions. J Cutan Pathol 2018;45:764–73. [DOI] [PubMed] [Google Scholar]
  • 86.Davis MD, Dinneen AM, Landa N, Gibson LE. Grover’s disease: clinicopathologic review of 72 cases. Mayo Clinic proceedings 1999;74:229–34. [DOI] [PubMed] [Google Scholar]
  • 87.Trinidad C, Nelson KC, Glitza Oliva IC, Torres-Cabala CA, Nagarajan P, Tetzlaff MT et al. Dermatologic toxicity from immune checkpoint blockade therapy with an interstitial granulomatous pattern. J Cutan Pathol 2018;45:504–7. [DOI] [PubMed] [Google Scholar]
  • 88.Eckert A, Schoeffler A, Dalle S, Phan A, Kiakouama L, Thomas L. Anti-CTLA4 monoclonal antibody induced sarcoidosis in a metastatic melanoma patient. Dermatology 2009;218:69–70. [DOI] [PubMed] [Google Scholar]
  • 89.Reule RB, North JP. Cutaneous and pulmonary sarcoidosis-like reaction associated with ipilimumab. J Am Acad Dermatol 2013;69:e272–e3. [DOI] [PubMed] [Google Scholar]
  • 90.Tissot C, Carsin A, Freymond N, Pacheco Y, Devouassoux G. Sarcoidosis complicating anti-cytotoxic T-lymphocyte-associated antigen-4 monoclonal antibody biotherapy. The European respiratory journal 2013;41:246–7. [DOI] [PubMed] [Google Scholar]
  • 91.Firwana B, Ravilla R, Raval M, Hutchins L, Mahmoud F. Sarcoidosis-like syndrome and lymphadenopathy due to checkpoint inhibitors. Journal of oncology pharmacy practice : official publication of the International Society of Oncology Pharmacy Practitioners 2017;23:620–4. [DOI] [PubMed] [Google Scholar]
  • 92.Tetzlaff MT, Nelson KC, Diab A, Staerkel GA, Nagarajan P, Torres-Cabala CA et al. Granulomatous/sarcoid-like lesions associated with checkpoint inhibitors: a marker of therapy response in a subset of melanoma patients. Journal for immunotherapy of cancer 2018;6:14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Birnbaum MR, Ma MW, Fleisig S, Packer S, Amin BD, Jacobson M et al. Nivolumab-related cutaneous sarcoidosis in a patient with lung adenocarcinoma. JAAD Case Rep 2017;3:208–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Danlos FX, Pages C, Baroudjian B, Vercellino L, Battistella M, Mimoun M et al. Nivolumab-Induced Sarcoid-Like Granulomatous Reaction in a Patient With Advanced Melanoma. Chest 2016;149:e133–6. [DOI] [PubMed] [Google Scholar]
  • 95.Lomax AJ, McGuire HM, McNeil C, Choi CJ, Hersey P, Karikios D et al. Immunotherapy-induced sarcoidosis in patients with melanoma treated with PD-1 checkpoint inhibitors: Case series and immunophenotypic analysis. International journal of rheumatic diseases 2017;20:1277–85. [DOI] [PubMed] [Google Scholar]
  • 96.Selim A, Ehrsam E, Atassi MB, Khachemoune A. Scar sarcoidosis: a case report and brief review. Cutis 2006;78:418–22. [PubMed] [Google Scholar]
  • 97.Kim YC, Triffet MK, Gibson LE. Foreign bodies in sarcoidosis. Am J Dermatopathol 2000;22:408–12. [DOI] [PubMed] [Google Scholar]
  • 98.Hwang SJ, Carlos G, Wakade D, Sharma R, Fernandez-Penas P. Ipilimumab-induced acute generalized exanthematous pustulosis in a patient with metastatic melanoma. Melanoma Res 2016;26:417–20. [DOI] [PubMed] [Google Scholar]
  • 99.Gormley R, Wanat K, Elenitsas R, Giles J, McGettigan S, Schuchter L et al. Ipilimumab-associated Sweet syndrome in a melanoma patient. J Am Acad Dermatol 2014;71:e211–3. [DOI] [PubMed] [Google Scholar]
  • 100.Kyllo RL, Parker MK, Rosman I, Musiek AC. Ipilimumab-associated Sweet syndrome in a patient with high-risk melanoma. J Am Acad Dermatol 2014;70:e85–6. [DOI] [PubMed] [Google Scholar]
  • 101.Pintova S, Sidhu H, Friedlander PA, Holcombe RF. Sweet’s syndrome in a patient with metastatic melanoma after ipilimumab therapy. Melanoma Res 2013;23:498–501. [DOI] [PubMed] [Google Scholar]
  • 102.Rudolph BM, Staib F, Von Stebut E, Hainz M, Grabbe S, Loquai C. Neutrophilic disease of the skin and intestines after ipilimumab treatment for malignant melanoma -simultaneous occurrence of pyoderma gangrenosum and colitis. Eur J Dermatol 2014;24:268–9. [DOI] [PubMed] [Google Scholar]
  • 103.Wu BC, Patel ED, Ortega-Loayza AG. Drug-induced pyoderma gangrenosum: a model to understand the pathogenesis of pyoderma gangrenosum. Br J Dermatol 2017;177:72–83. [DOI] [PubMed] [Google Scholar]
  • 104.Carlos G, Anforth R, Chou S, Clements A, Fernandez-Penas P. A case of bullous pemphigoid in a patient with metastatic melanoma treated with pembrolizumab. Melanoma Res 2015;25:265–8. [DOI] [PubMed] [Google Scholar]
  • 105.Naidoo J, Schindler K, Querfeld C, Busam K, Cunningham J, Page DB et al. Autoimmune Bullous Skin Disorders with Immune Checkpoint Inhibitors Targeting PD-1 and PD-L1. Cancer immunology research 2016;4:383–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Damsky W, Kole L, Tomayko MM. Development of bullous pemphigoid during nivolumab therapy. JAAD Case Rep 2016;2:442–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Kwon CW, Land AS, Smoller BR, Scott G, Beck LA, Mercurio MG. Bullous pemphigoid associated with nivolumab, a programmed cell death 1 protein inhibitor. J Eur Acad Dermatol Venereol 2017;31:e349–e50. [DOI] [PubMed] [Google Scholar]
  • 108.Rofe O, Bar-Sela G, Keidar Z, Sezin T, Sadik CD, Bergman R. Severe bullous pemphigoid associated with pembrolizumab therapy for metastatic melanoma with complete regression. Clin Exp Dermatol 2017;42:309–12. [DOI] [PubMed] [Google Scholar]
  • 109.Sowerby L, Dewan AK, Granter S, Gandhi L, LeBoeuf NR. Rituximab Treatment of Nivolumab-Induced Bullous Pemphigoid. JAMA Dermatol 2017;153:603–5. [DOI] [PubMed] [Google Scholar]
  • 110.Russo I, Sacco G, Frega S, Polo V, Pasello G, Alaibac M. Immunotherapy-related skin toxicity: bullous pemphigoid in a lung adenocarcinoma patient treated with the anti-PDL1 antibody atezolizumab. Eur J Dermatol 2017;27:205–8. [DOI] [PubMed] [Google Scholar]
  • 111.Mochel MC, Ming ME, Imadojemu S, Gangadhar TC, Schuchter LM, Elenitsas R et al. Cutaneous autoimmune effects in the setting of therapeutic immune checkpoint inhibition for metastatic melanoma. J Cutan Pathol 2016;43:787–91. [DOI] [PubMed] [Google Scholar]
  • 112.Kasperkiewicz M, Zillikens D. The pathophysiology of bullous pemphigoid. Clinical reviews in allergy & immunology 2007;33:67–77. [DOI] [PubMed] [Google Scholar]
  • 113.Xia Y, Jeffrey Medeiros L, Young KH. Signaling pathway and dysregulation of PD1 and its ligands in lymphoid malignancies. Biochim Biophys Acta 2016;1865:58–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Petukhova L, Duvic M, Hordinsky M, Norris D, Price V, Shimomura Y et al. Genome-wide association study in alopecia areata implicates both innate and adaptive immunity. Nature 2010;466:113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.John KK, Brockschmidt FF, Redler S, Herold C, Hanneken S, Eigelshoven S et al. Genetic variants in CTLA4 are strongly associated with alopecia areata. J Invest Dermatol 2011;131:1169–72. [DOI] [PubMed] [Google Scholar]
  • 116.Carroll JM, McElwee KJ, L EK, Byrne MC, Sundberg JP. Gene array profiling and immunomodulation studies define a cell-mediated immune response underlying the pathogenesis of alopecia areata in a mouse model and humans. J Invest Dermatol 2002;119:392–402. [DOI] [PubMed] [Google Scholar]
  • 117.Zarbo A, Belum VR, Sibaud V, Oudard S, Postow MA, Hsieh JJ et al. Immune-related alopecia (areata and universalis) in cancer patients receiving immune checkpoint inhibitors. Br J Dermatol 2017;176:1649–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Rivera N, Boada A, Bielsa MI, Fernandez-Figueras MT, Carcereny E, Moran MT et al. Hair Repigmentation During Immunotherapy Treatment With an Anti-Programmed Cell Death 1 and Anti-Programmed Cell Death Ligand 1 Agent for Lung Cancer. JAMA Dermatol 2017;153:1162–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Uenami T, Hosono Y, Ishijima M, Kanazu M, Akazawa Y, Yano Y et al. Vitiligo in a patient with lung adenocarcinoma treated with nivolumab: A case report. Lung cancer (Amsterdam, Netherlands) 2017;109:42–4. [DOI] [PubMed] [Google Scholar]
  • 120.Yin ES, Totonchy MB, Leventhal JS. Nivolumab-associated vitiligo-like depigmentation in a patient with acute myeloid leukemia: A novel finding. JAAD Case Rep 2017;3:90–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Hua C, Boussemart L, Mateus C, Routier E, Boutros C, Cazenave H et al. Association of Vitiligo With Tumor Response in Patients With Metastatic Melanoma Treated With Pembrolizumab. JAMA Dermatol 2016;152:45–51. [DOI] [PubMed] [Google Scholar]
  • 122.Larsabal M, Marti A, Jacquemin C, Rambert J, Thiolat D, Dousset L et al. Vitiligo-like lesions occurring in patients receiving anti-programmed cell death-1 therapies are clinically and biologically distinct from vitiligo. J Am Acad Dermatol 2017;76:863–70. [DOI] [PubMed] [Google Scholar]
  • 123.Houghton AN, Eisinger M, Albino AP, Cairncross JG, Old LJ. Surface antigens of melanocytes and melanomas. Markers of melanocyte differentiation and melanoma subsets. J Exp Med 1982;156:1755–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Staser K, Chen D, Solus J, Rosman IS, Schaffer A, Cornelius L et al. Extensive tumoral melanosis associated with ipilimumab-treated melanoma. Br J Dermatol 2016;175:391–3. [DOI] [PubMed] [Google Scholar]
  • 125.Libon F, Arrese JE, Rorive A, Nikkels AF. Ipilimumab induces simultaneous regression of melanocytic naevi and melanoma metastases. Clin Exp Dermatol 2013;38:276–9. [DOI] [PubMed] [Google Scholar]
  • 126.Yuan J, Ginsberg B, Page D, Li Y, Rasalan T, Gallardo HF et al. CTLA-4 blockade increases antigen-specific CD8(+) T cells in prevaccinated patients with melanoma: three cases. Cancer immunology, immunotherapy : CII 2011;60:1137–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Freites-Martinez A, Kwong BY, Rieger KE, Coit DG, Colevas AD, Lacouture ME. Eruptive Keratoacanthomas Associated With Pembrolizumab Therapy. JAMA dermatology 2017;153:694–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Feldstein SI PF, Larsen L, Kim E, Hwang S, Fung MA. Eruptive keratoacanthomas arising in the setting of lichenoid toxicity after programmed cell death 1 inhibition with nivolumab. Journal of the European Academy of Dermatology and Venereology : JEADV 2018;32:e58–e9. [DOI] [PubMed] [Google Scholar]
  • 129.Bousquet E, Zarbo A, Tournier E, Chevreau C, Mazieres J, Lacouture ME et al. Development of Papulopustular Rosacea during Nivolumab Therapy for Metastatic Cancer. Acta Derm Venereol 2017;97:539–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Shenoy N, Esplin B, Barbosa N, Wieland C, Thanarajasingam U, Markovic S. Pembrolizumab induced severe sclerodermoid reaction. Annals of oncology : official journal of the European Society for Medical Oncology 2017;28:432–3. [DOI] [PubMed] [Google Scholar]
  • 131.Sheik Ali S, Goddard AL, Luke JJ, Donahue H, Todd DJ, Werchniak A et al. Drug-associated dermatomyositis following ipilimumab therapy: a novel immune-mediated adverse event associated with cytotoxic T-lymphocyte antigen 4 blockade. JAMA Dermatol 2015;151:195–9. [DOI] [PubMed] [Google Scholar]
  • 132.Yamaguchi Y, Abe R, Haga N, Shimizu H. A case of drug-associated dermatomyositis following ipilimumab therapy. Eur J Dermatol 2016;26:320–1. [DOI] [PubMed] [Google Scholar]
  • 133.Sibaud V, David I, Lamant L, Resseguier S, Radut R, Attal J et al. Acute skin reaction suggestive of pembrolizumab-induced radiosensitization. Melanoma Res 2015;25:555–8. [DOI] [PubMed] [Google Scholar]
  • 134.Mauzo SH, Tetzlaff MT, Nelson K, Amaria R, Patel S, Aung PP et al. Regressed melanocytic nevi secondary to pembrolizumab therapy: an emerging melanocytic dermatologic effect from immune checkpoint antibody blockade. International journal of dermatology 2019;58:1045–52. [DOI] [PubMed] [Google Scholar]

RESOURCES