Abstract
Background:
Severe cutaneous adverse reactions (SCARs) are associated with high morbidity and mortality in cancer patients. Early identification and treatment of SCARs may improve outcomes.
Objective:
To identify biomarkers to predict outcomes in hospitalized cancer patients who developed SCARs.
Methods:
Retrospective review of 144 hospitalized cancer patients with a morbilliform rash, recorded testing for serum cytokines (IL-6, IL-10, and TNF-α) or elafin, and a dermatology consultation. Rashes were categorized as ‘simple’ morbilliform rash without systemic involvement or ‘complex’ morbilliform rash with systemic involvement.
Results:
Fifty-four of 144 (37.5%) patients died during follow-up. Elevated levels of IL-6, IL-10, and TNF-α were associated with decreased survival. OS in patients with elevated levels of IL-6, IL-10, and TNF-α was 53.7%, 56.6%, 53.6%, respectively compared to 85.7%, 82.5% and 83.6% with lower levels. Patients with increased levels of both IL-6 and TNF-α had a nearly 6-fold increase in mortality (HR 5.82) compared to patients with lower levels.
Limitations:
Retrospective design, limited sample size, and high-risk population.
Conclusions:
Hospitalized cancer patients with rash and elevated IL-6 and TNF-α were nearly 6 times more likely to die over the course of follow-up. These biomarkers may serve as prognostic biomarkers and therapeutic targets for this high-risk population.
Keywords: Severe cutaneous adverse reaction, cytokine, biomarker, drug reaction, drug reaction with eosinophilia and systemic symptoms, drug induced hypersensitivity syndrome, graft versus host disease, drug rash, interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α), mortality, survival
Capsule Summary
Reliable prognostic biomarkers are needed to predict outcomes in hospitalized cancer patients with rash.
Hospitalized cancer patients with rash and elevated IL-6 and TNF-α levels have a nearly 6 times decreased overall survival. Given the availability of TNF-α and IL-6 inhibitors, these biomarkers may serve as potential therapeutic targets.
Introduction
Patients with cancer are at risk of developing therapy-related morbilliform eruptions, graft-versus-host disease (GVHD), and severe cutaneous adverse reactions (SCARs), including Stevens-Johnson syndrome (SJS), toxic epidermal necrolysis (TEN), and drug-induced hypersensitivity (DIHS)/drug reaction with eosinophilia and systemic symptoms (DRESS). SCARs are 28–57 times more common in cancer patients, with 5.7–14.9 cases per 100,000 patients, and more fatal (mortality of 32% with vs. 8.5% without cancer in SJS/TEN) with the greatest risk among hematologic cancer patients1–3. The increased mortality in this population is thought to be multifactorial due to malnutrition, cancer type, immunocompromised status, and chemotherapy type.4
Early recognition and treatment of SCARs is vital for improving survival.4 Earlier withdrawal of the causative drug is associated with better SCAR prognosis (odds ratio, 0.69 for each day; 95% CI: 0.53–0.89).5 Given the increased incidence and mortality of SCARs in cancer patients, and the difficulty in clinically diagnosing SCARs, reliable, objective markers are needed. Our previous study found an association between elafin, interleukin-6 (IL-6), and tumor necrosis factor alpha (TNF-α) with all-cause mortality in cancer patients who develop SCARs and interleukin-10 (IL-10) and IL-6 with a drug-related complex rash.6 Elafin is overexpressed in wound healing and inflammatory skin disorders and is a diagnostic and prognostic plasma biomarker in GVHD. Elevated IL-6 and IL-10 levels have been found in patients with acute GVHD, SJS, TEN.7, 8
Biomarkers have potential prognostic and diagnostic utility in a hospitalized cancer population who develop SCARs. The objective of this study was to identify serologic markers or combinations of markers that may be used to predict outcomes in hospitalized cancer patients who developed SCARs.
Methods
This was a retrospective cohort study approved by the Institutional Review Board of Memorial Sloan Kettering Cancer Center (MSKCC). A database query of adult patients with cancer who were hospitalized between 8/2016–2/2019 and had ICD 9 or 10 codes for rash (R23, R21, 693, 692, 695, 690–698, L20-L30, L51, L43.2, T88.7, L55–59), recorded testing for serum biomarkers IL-1β, IL-6, IL-10, TNF-α, or plasma elafin, and prior dermatology consultation revealed 151 eligible patients (Figure 1). These biomarkers are obtained at our institution as standard of care for all patients who present with possible drug eruption to complement the clinical picture and serve as a potential therapeutic target. Seven patients were excluded because biomarkers were checked for a reason other than a morbilliform rash (i.e., cytokine release syndrome, study protocol, sepsis, or cellulitis/panniculitis).
Figure 1:
Flowchart of patient selection. GVHD, Graft-versus-host disease; ICD, International Classification of Diseases; MSKCC, Memorial Sloan Kettering Cancer Center; 2/2, secondary to
One hundred and forty-four patients were evaluated on the inpatient service or in the urgent care center at MSKCC with a diagnosis of morbilliform rash and tested for cytokines or elafin. Chart review was performed for all patients to assign ‘simple’ and ‘complex’ morbilliform rash groups. ‘Simple’ morbilliform rash was defined as a rash with no systemic involvement, or spontaneous resolution of rash with remote systemic involvement, or limited course of rash that did not require systemic therapy. ‘Complex’ morbilliform rash was defined as a SCAR with systemic organ involvement requiring systemic therapy with a prolonged course.
For each patient, a modified RegiSCAR score9 was calculated based on the following items: fever ≥ 38.5°C (0 points, −1 if absent); peripheral eosinophilia (≥ 700/mm3 or ≥ 10%; 1 point or ≥ 1500/mm3 or ≥ 20%; 2 points); presence of atypical lymphocytes (1 point); rash ≥ 50% of body surface area (1 point) with facial edema, purpura, infiltration, or desquamation (1 point); organ involvement (1 point for one organ, 2 points for two or more); disease duration > 15 days (0 points, −1 if absent); at least 3 biological investigations (e.g., blood cultures, viral serology, biopsy) performed and negative to rule out an alternative diagnosis (1 point). Comprehensive metabolic panel, including glomerular filtration rate (GFR), blood urea nitrogen (BUN), creatinine (Cr), transaminases, total bilirubin and urine eosinophils, were reviewed. For all laboratory values, only results within 7 days of biomarker testing were recorded to minimize the impact of non-rash events on biomarker levels. Reference values for cytokines (IL-10 ≤18 pg/mL, IL-6 ≤5 pg/mL, TNF-α ≤22 pg/mL) are determined by our institution’s laboratory, while that for elafin (≤23.8 ng/mL) is determined by Viracor, Inc.
Descriptive statistics and graphical methods were used to assess distributions of patient and medical test characteristics. Chi-square tests and Fisher’s Exact test were used to assess the association between rash type (simple versus complex) and nominally scaled patient and medical test characteristics. T-tests and Wilcoxon rank-sum tests were used to assess differences in continuously scaled variables by rash type. Overall survival (OS) was calculated as the time elapsed between initial assessment for SCARs to death from any cause. A patient was considered censored at the date of last follow-up date if she or he were alive at that point of contact. Kaplan-Meier estimates with log rank tests were used to assess probabilities of survival. Cox proportional hazards regression models were used to explore prognostic factors associated with survival while controlling for patient characteristics. Proportionality assumptions for Cox models were evaluated by the visual assessment of Cox-Snell and standardized residuals. All p-values are two-sided. Analyses were performed with STATA 16.0 software (StataCorp LP, College Station, TX).
Results
Patient characteristics
One hundred and forty-four patients with cancer and morbilliform rash who were evaluated at a single institution for dermatology consultation were included in the study. Of these patients, 50.6% were male, and the average age was 55.5 years. Of the 144 patients, 81 (56.3%) had a ‘simple’ rash and 63 (43.7%) had a ‘complex’ rash with systemic involvement. Among complex rash patients, 46% (n=29) had cutaneous graft-versus-host disease (GVHD), 6.3% (n=4) had cutaneous GVHD-spectrum rash (engraftment syndrome), and the remaining 46.9% (n=30) had a complex rash secondary to drug exposure (Table I). Most patients with complex rash had hematologic malignancy (69.8%), while most patients with a simple rash had solid malignancy (56.8%).
Table I.
Characteristics of hospitalized cancer patients and ‘simple’ morbilliform rash vs. ‘complex’ morbilliform rash. GVHD, Graft-versus-host disease; BSA, Body surface area; CTCAE, Common Terminology Criteria for Adverse Events; GFR, glomerular filtration rate; SCAR, severe cutaneous adverse reaction.
Category | Rash type | Total | p-value | ||
---|---|---|---|---|---|
Simple morbilliform rash, n=81, n (%) | Complex morbilliform rash, n=63), n (%) | ||||
Patient Sex | Female | 40 (49.4) | 31 (49.2) | 71 (49.3) | 0.983 |
Male | 41 (50.6) | 32 (50.8) | 73 (50.7) | ||
Location | Inpatient | 49 (60.5) | 53 (84.1) | 102 (70.8) | 0.002 |
Urgent care center | 32 (39.5) | 10 (15.9) | 42 (29.2) | ||
Cancer type | Solid | 46 (56.8) | 18 (28.6) | 64 (44.4) | 0.001 |
Hematologic | 32 (39.5) | 44 (69.8) | 76 (52.8) | ||
Both | 3 (3.7) | 1 (1.6) | 4 (2.8) | ||
Diagnosis | complex drug | 1 (1.2) | 30 (47.6) | 31 (21.5) | <0.001 |
GVHD | 0 (0) | 29 (46) | 29 (20.1) | ||
simple drug | 77 (95.1) | 0 (0) | 77 (53.5) | ||
GVHD-like | 0 (0) | 4 (6.4) | 4 (2.8) | ||
viral exanthem | 3 (3.7) | 0 (0) | 3 (2.1) | ||
CTCAE Grade | 0 | 0 (0) | 1 (1.6) | 1 (0.7) | 0.002 |
1 | 9 (11.1) | 0 (0) | 9 (6.3) | ||
2 | 28 (34.6) | 13 (20.6) | 41 (28.5) | ||
3 | 44 (54.3) | 46 (73) | 90 (62.5) | ||
4 | 0 (0) | 3 (4.8) | 3 (2.1) | ||
RegiSCAR score | 0 | 33 (40.7) | 3 (4.8) | 36 (25) | <0.001 |
1 | 28 (34.6) | 14 (22.2) | 42 (29.2) | ||
2 | 16 (19.8) | 14 (22.2) | 30 (20.8) | ||
3 | 3 (3.7) | 24 (38.1) | 27 (18.8) | ||
4 | 1 (1.2) | 7 (11.1) | 8 (5.6) | ||
5 | 0 (0) | 1 (1.6) | 1 (0.7) | ||
Atypical | No | 78 (96.3) | 54 (87.1) | 132 (92.3) | 0.041 |
Lymphocytes | Yes | 3 (3.7) | 8 (12.9) | 11 (7.7) | |
Rash, (50% BSA + purpura, edema, scale) | 0 | 37 (46.3) | 3 (4.8) | 40 (28) | <0.001 |
1 | 30 (37.5) | 29 (46) | 59 (41.3) | ||
2 | 13 (16.3) | 31 (49.2) | 44 (30.8) | ||
Fever, >38°C | No | 5 (6.2) | 8 (12.7) | 13 (9) | 0.175 |
Yes | 76 (93.8) | 55 (87.3) | 131 (91) | ||
Decreased GFR relative to baseline | No | 72 (90) | 47 (74.6) | 119 (83.2) | 0.04 |
Yes | 8 (10) | 16 (25.4) | 24 (16.8) | ||
Elevated transaminases relative to baseline | No | 68 (84) | 28 (44.4) | 96 (66.7) | <0.001 |
Yes | 13 (16.1) | 35 (55.6) | 48 (33.3) | ||
Elevated total bilirubin relative to baseline | No | 76 (93.8) | 55 (87.3) | 131 (91) | 0.175 |
Yes | 5 (6.2) | 8 (12.7) | 13 (9) | ||
Internal organs involved | 0 | 59 (72.8) | 12 (19.1) | 71 (49.3) | <0.001 |
1 | 22 (27.2) | 43 (68.3) | 65 (45.1) | ||
2 | 0 (0) | 8 (12.7) | 8 (5.6) | ||
Respiratory virus panel positive | No | 43 (86) | 46 (93.9) | 89 (89.9) | 0.193 |
Yes | 7 (14) | 3 (6.1) | 10 (10.1) | ||
Human herpesvirus positive | No | 36 (87.8) | 41 (82) | 77 (84.6) | 0.445 |
Yes | 5 (12.2) | 9 (18) | 14 (15.4) | ||
Cytomegalovirus positive | No | 42 (93.3) | 48 (90.6) | 90 (91.8) | 0.618 |
Yes | 3 (6.7) | 5 (9.4) | 8 (8.2) | ||
Epstein-Barr virus positive | No | 41 (93.2) | 52 (92.9) | 93 (93) | 0.950 |
Yes | 3 (6.8) | 4 (7.1) | 7 (7) | ||
Adenovirus positive | No | 40 (100) | 48 (98) | 88 (98.9) | 0.364 |
Yes | 0 (0) | 1 (2) | 1 (1.1) | ||
Skin biopsy supportive of drug reaction | No | 56 (69.1) | 32 (50.8) | 88 (61.1) | 0.025 |
Yes | 25 (30.9) | 31 (49.2) | 56 (38.9) | ||
Status | Alive | 53 (65.4) | 35 (55.6) | 88 (61.1) | 0.228 |
Dead | 28 (34.6) | 28 (44.4) | 56 (38.9) |
Biomarkers
The median values of cytokines (IL-10, IL-6, TNF-α) and elafin were higher in the complex rash patients compared to those with simple rash, although only IL-10 reached statistical significance (p=0.03) (Table II). Peripheral eosinophilia and white blood cell count did not differ between simple and complex rash groups.
Table II.
Comparison of median values of patient characteristics by ‘simple’ morbilliform rash vs. ‘complex’ morbilliform rash. IL, interleukin; TNF-α, tumor necrosis factor α
Characteristic | Overall Median | Rash type | Median | Binomial Interpolation of 95% CI | p-value | |
---|---|---|---|---|---|---|
Lower | Upper | |||||
Age (years) | 60.5 | Simple | 62 | 56.2 | 66.8 | 0.17 |
Complex | 56 | 53.0 | 63.0 | |||
IL-1b | 0 | Simple | 0 | 0 | 0 | 0.38 |
Complex | 0 | 0 | 0 | |||
IL-10 | 17.5 | Simple | 14 | 10.0 | 19.5 | 0.03 |
Complex | 26.5 | 16.0 | 31.6 | |||
IL-6 | 24.5 | Simple | 17.5 | 10.5 | 30.0 | 0.14 |
Complex | 31.5 | 22.8 | 50.8 | |||
TNF-α | 14 | Simple | 12.5 | 10.0 | 16.5 | 0.27 |
Complex | 16 | 13.4 | 19.0 | |||
Elafin | 18.35 | Simple | 17 | 13.3 | 23.3 | 0.43 |
Complex | 20.2 | 17.2 | 28.0 | |||
% Eosinophils | 1.95 | Simple | 2.1 | 1.0 | 2.8 | 0.80 |
Complex | 1.8 | 0.2 | 4.7 | |||
Absolute eosinophils | 0.1 | Simple | 0.1 | 0.1 | 0.2 | 0.96 |
Complex | 0.15 | 0 | 0.3 | |||
White blood cells | 5.65 | Simple | 5.5 | 4.6 | 6.7 | 0.72 |
Complex | 5.7 | 4.3 | 7.8 |
Biomarkers and organ involvement
The median IL-10 level was significantly higher in patients with elevated transaminases than in patients with transaminases in the normal range (24.5 pg/mL vs. 14 pg/mL, p= 0.01). Median IL-6 levels were also elevated in patients with elevated transaminases, however this difference did not reach statistical significance (41 pg/mL vs 19.5 pg/mL, p=0.056). There was no significant association between IL-10, IL-6, TNF-α, or elafin and bilirubin or renal involvement, as measured by decreased glomerular filtration rate relative to baseline.
Overall survival
Median follow-up time for the cohort was 14.7 months. Fifty-four of 144 (37.5%) patients died during follow-up. Six-month OS was not statistically significantly different between complex versus simple rash, or between patients with a Common Terminology Criteria for Adverse Events (CTCAE) v5.0 score of 0–2 versus 3–4. Patients with morbilliform rash and elevated IL-6 levels had a decreased OS that worsened in concordance with rising IL-6 levels. At 6 months, patients with IL-6 levels above the median (>24 pg/mL) had an OS of 53.7% (95% CI: 39.7–65.5), compared to 85.7% OS (95% CI: 73.5–92.6) in those with lower IL-6 levels (≤ 24pg/mL), p<0.001. Patients with TNF-α levels above the median value (>14 pg/mL) had a significantly shorter OS at 6 months than those with TNF-α levels in the bottom two quartiles (≤14 pg/mL), 56.6% versus 82.5%, p<0.001. Elevated IL-10 levels were also associated with decreased 6-month OS, as patients with IL-10 levels greater than the median (≥18 pg/ml) had a 6-month survival of 53.6% (95% CI: 42.5–68.0) compared to 83.6% (95% CI: 70.9– 91.1) for those below the median, p=0.006. Table III presents univariate and multivariate Cox models for OS. In univariate analysis, higher levels of IL-6, IL-10 and TNF-α were all associated with worse prognosis. On multivariate analysis examining cytokines, rash type (simple versus complex) and patient age, patients with elevated levels of IL-6 were 3.2 times more likely to die (HR = 3.21, 95% CI: 1.64–6.32) over the course of the follow-up compared to those with lower levels. Similarly, patients with higher levels of TNF-α were also more likely to die compared to those with lower levels (HR = 1.78, 95% CI 0.94– 3.35). Figure 2 depicts the multivariate survival experience of patients with the combination of increased levels of IL-6 and TNF-α. These patients had a 5-fold increase in mortality compared to those with lower levels of both measures (HR =5.82, 95% CI: 1.53– 17.43). No difference was observed in 6-month overall survival in cancer patients with simple versus complex rash and elevated absolute eosinophils.
Table III.
Univariate and multivariate analyses of Overall Survival using Cox proportional hazards models. IL, interleukin; TNF-α, tumor necrosis factor α
Category | Univariate | Multivariate* | ||||
---|---|---|---|---|---|---|
HR for death (95% CI) | p-value | HR for death (95% CI) | p-value | |||
Rash Type | Simple | referent | 0.165 | referent | 0.666 | |
Complex | 1.46 (0.85– 2.51) | 0.88 (0.48– 1.59) | ||||
IL-6 | 0–24 | referent | <0.001 | referent | 0.001 | |
25/max | 3.77 (2.02–7.04) | 3.21 (1.64– 6.32) | ||||
IL-10 | min/17 | referent | 0.008 | referent | 0.386 | |
18/max | 2.22 (1.23– 4.02) | 1.35 (0.68– 2.70) | ||||
TNF-α | min/14 | referent | 0.001 | referent | 0.076 | |
15/max | 2.66 (1.46– 4.84) | 1.78 (0.94– 3.35) |
Variables included in multivariate are the ones listed in the table, along with age.
Figure 2.
Survival functions for patients with IL-6 and TNF-α above vs. below the median. In both groups, the model predictions are based on patients with a mean age of 55.5 years. Variables included in the multivariate analyses are IL-6, TNF-α, IL-10, rash type (simple morbilliform vs complex morbilliform), and age. IL, interleukin; TNF-α, tumor necrosis factor α
Discussion
This study examined patients with cancer who presented with morbilliform rash and found the combination of elevated TNF-α and IL-6 to be significantly associated with a nearly 6 times increased all-cause mortality after rash onset (p<0.001). While past studies have shown an increased mortality associated with elevated IL-10, IL-6, and elafin, the prognostic value of combinations of cytokines have not been elucidated.6, 10 Using multiple biomarkers as opposed to single cytokines can strengthen the overall prognostic value and serve as a reliable tool for SCAR diagnosis and prognostication.11 Our study did not find a statistically significant difference in OS at 6 months between patients with complex and simple rashes (69.5% compared to 75.8%, respectively, p=0.23), highlighting the need for objective tools to guide patients who require therapy. The availability of TNF-α and IL-6 inhibitors make these biomarkers attractive therapeutic targets for intervention. The Food and Drug Administration’s approval of the IL-6 receptor antibody tocilizumab for the treatment of cytokine release syndrome (CRS) further underscores the therapeutic potential of identifying inflammatory markers in supportive onco-dermatology.12 The severity of symptoms in CRS may correlate with duration of exposure to the inflammatory cytokines, emphasizing the importance of prompt recognition of these markers for both diagnostic and treatment purposes.13
Our study found that levels of IL-6 in cancer patients with morbilliform rash are inversely related to OS, confirming in a larger cohort the previous report by Mori et al.6 IL-6 is a pleiotropic cytokine with pro-inflammatory effects that inhibit the induction of regulatory T-cells by TGF-beta and shifts naïve T-cell differentiation towards Th17 cells.14, 15 Th17 cells release large quantities of IL-17, IL-22, and IL-21, and is a major inducer of tissue inflammation.16 Significant elevations in serum IL-6 have previously been reported in patients with SCARs and have also been associated with increased incidence and severity of GVHD.6, 11, 17 IL-6 is implicated in the development of rashes and elevated lesional IL-6 levels have been found in numerous other inflammatory skin disorders .18–20 Administration of recombinant IL-6 to a patient resulted in an inflammatory skin eruption with recurrence after rechallenge.21 Although the exact mechanism is not well understood, the rapid resolution of rash in CRS with the use of IL-6 inhibitors implies the pathophysiologic importance of IL-6.12 Moreover, IL-6 mediates tumor progression and therapeutic resistance.22–24 Elevated levels of IL-6 in cancer patients have been associated with poor outcomes and addition of an IL-6 blockade to immune checkpoint inhibitor therapy and mRNA-based immunotherapy has resulted in a significant reduction in tumor volume and led to prolonged survival in mice.22, 25 IL-6 blockade reversed anti-PD-L1 resistance and prolonged tumor-bearing mouse survival in a preclinical colorectal cancer model.26 IL-6 blockade also augments regulatory T cell reconstitution.17 IL-6 inhibitors have been effectively used in GVHD prophylaxis and treatment of steroid-refractory acute and chronic GVHD.27–29 IL-6 blockade receptors in murine models of GVHD significantly reduced GVHD-associated mortality.30 Given our findings of decreased OS in cancer patients with rash and high IL-6, IL-6 may represent an actionable therapeutic target to improve OS in this patient population.
TNF-α, a pro-inflammatory cytokine, appears to be involved in the pathogenesis of severe cutaneous adverse reactions. TNF-α induces keratinocyte apoptosis through a Fas-mediated pathway, leading to the extensive epithelial cell death seen in SJS/TEN.31 This mechanism of action may explain the elevated TNF-α frequently found in the plasma and blister fluids of patients with SJS/TEN, and in the plasma of patients with GVHD and acute generalized exanthematous pustulosis (AGEP).7, 32–35 Additionally, TNF-α has been linked to promotion and progression of cancer.36, 37 Levels of IL-6 and TNF-α directly correlated with the extent of disease in patients with prostate cancer.38 TNF-α was also an independent predictor of poor survival in cancer patients, highlighting the potential role of TNF-α antagonists in the treatment of cancer patients.39, 40 Furthermore, TNF-α inhibitors are an effective treatment for steroid-refractory aGVHD and SCARs.32, 41–43 Moreover, a randomized controlled trial found that TNF-α antagonists decrease the predicted mortality rate and healing time in patients with cytotoxic T lymphocyte-mediated SCARs (including SJS-TEN).44
In our study, elevated IL-10 levels in cancer patients with morbilliform rash were significantly associated with subsequent development of a complex rash (p=0.03) and decreased OS at 6 months (p=0.006). IL-10 is an anti-inflammatory cytokine produced by Th2 cells and keratinocytes in the skin that inhibits the production of IL-6 and TNF-α.45, 46 IL-10 may act as a defense mechanism against excessive tissue inflammation.47 Patients with SJS/TEN may have elevated blister fluid concentrations of IL-10.7Past studies have similarly found that elevated IL-10 levels may be produced as a result of excessive tissue inflammation in toxic epidermal necrolysis and reflect a compensatory response.46 Elevated levels of IL-10 have also been correlated with a poor response in numerous types of cancer, however the role of IL-10 in tumorigenesis remains controversial.48–50 Its immunosuppressive influence has been thought to reduce the anti-tumor immune response, yet murine tumor models have demonstrated rapid tumor rejection with increased IL-10 secretion.51 Given the significantly decreased 6-month OS (p=0.006) in patients with elevated IL-10 levels in our cohort, our study supports prior literature in cancer patients and suggests that this marker may also have prognostic value in cancer patients at the time of rash development.49, 52
Although eosinophilia is a criterion on the RegiSCAR scoring system for identifying patients with DRESS/DIHS, there was no difference in 6-month overall survival in cancer patients with morbilliform rash and elevated absolute eosinophils. Past studies have similarly found that eosinophils are associated with immune-related adverse events but not overall survival.53
Limitations of this study include its retrospective design and limited sample size. All cases were recruited from a tertiary referral cancer center. A larger, prospective study examining the effect of IL-6 or TNF-α inhibitors on overall survival in patient with elevated IL-6 or TNF-α is needed. This exploratory analysis presents potential therapeutic targets to improve overall survival in a high-risk patient population.
Conclusion
This study highlights the prognostic significance of biomarker combinations for cancer patients with a morbilliform rash. The combination of elevated IL-6 and TNF-α in this population is significantly associated with a nearly 6 times decreased OS (P<0.001), while IL-10 may predict progression of a simple morbilliform rash to a SCAR. A growing number of studies suggest an increased survival associated with IL-6 and TNF-α inhibition in cancer or SCARs. Further studies evaluating the impact of TNF-α and IL-6 inhibitors on survival in this population are needed.
Funding sources:
Dr. Markova is supported by a Dermatology Foundation Career Development Award. This study was also funded in part by a grant from the National Cancer Institute / National Institutes of Health (P30-CA008748) made to the Memorial Sloan Kettering Cancer Center.
MEL: consultant/speaking role with Legacy Healthcare Services, ADC Therapeutics America Inc, Adgero Bio Pharmaceuticals, Amryt Pharmaceuticals, Apricity Health LLC, Azitra Inc, Celldex Therapeutics, Deciphera, Debiopharm, Galderma Research and Development, Johnson and Johnson, NCODA, Novocure Inc, Kyowa Kirin Inc, Lindi, Loxo, Merck Sharp and Dohme Corporation, Helsinn Healthcare SA, Janssen Research & Development LLC, Menlo Therapeutics, Novartis Pharmaceuticals Corporation, F. Hoffmann-La Roche AG, QED Therapeutics, AbbVie Inc, Boehringer Ingelheim Pharma Gmbh & Co. KG, Allergan Inc, Amgen Inc, E.R. Squibb & Sons LLC, EMD Serono Inc, Astrazeneca Pharmaceuticals LP, Genentech Inc, Leo Pharma Inc, Seattle Genetics, Bayer, Manner SAS, Lutris, Pierre Fabre, Paxman Coolers, Adjucare, Dignitana, Biotechspert, Teva Mexico, Parexel, OnQuality Pharmaceuticals Ltd, Takeda Millenium, and Our Brain Bank. Dr. Lacouture also receives research funding from Bristol-Myers Squibb, Lutris, Paxman, Novocure, US Biotest, and Veloce.
AM: advisory board of AstraZeneca. Dr. Markova also receives research funding from Incyte.
Abbreviations
- SCAR
severe cutaneous adverse reaction
- SJS
Stevens-Johnson syndrome
- TEN
toxic epidermal necrolysis
- DIHS
drug-induced hypersensitivity syndrome
- DRESS
drug reaction with eosinophilia and systemic syndrome
- GVHD
graft versus host disease
- IL-6
interleukin-6
- IL-10
interleukin-10
- TNF-α
tumor necrosis factor alpha
- GFR
glomerular filtration rate
- OS
Overall Survival
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.
IRB approval status: Approved by the Institutional Review Board of Memorial Sloan Kettering Cancer Center.
Conflicts of interest disclosure:
JRS, TSV, SM, SWD do not have any conflicts of interest to disclose.
References
- 1.Gillis NK, Hicks JK, Bell GC, Daly AJ, Kanetsky PA , McLeod HL. Incidence and Triggers of Stevens-Johnson Syndrome and Toxic Epidermal Necrolysis in a Large Cancer Patient Cohort. Journal of Investigative Dermatology 2017;137:2021–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Mockenhaupt M, Viboud C, Dunant A, Naldi L, Halevy S, Bouwes Bavinck JN et al. Stevens-Johnson syndrome and toxic epidermal necrolysis: assessment of medication risks with emphasis on recently marketed drugs. The EuroSCAR-study. The Journal of investigative dermatology 2008;128:35–44. [DOI] [PubMed] [Google Scholar]
- 3.Wu J, Lee YY, Su SC, Wu TS, Kao KC, Huang CC et al. Stevens-Johnson syndrome and toxic epidermal necrolysis in patients with malignancies. The British journal of dermatology 2015;173:1224–31. [DOI] [PubMed] [Google Scholar]
- 4.Duong TA, Valeyrie-Allanore L, Wolkenstein P , Chosidow O. Severe cutaneous adverse reactions to drugs. The Lancet 2017;390:1996–2011. [DOI] [PubMed] [Google Scholar]
- 5.Garcia-Doval I, LeCleach L, Bocquet H, Otero XL , Roujeau JC. Toxic epidermal necrolysis and Stevens-Johnson syndrome: does early withdrawal of causative drugs decrease the risk of death? Archives of dermatology 2000;136:323–7. [DOI] [PubMed] [Google Scholar]
- 6.Mori S, Hickey A, Dusza SW, Lacouture ME , Markova A. Markers of systemic involvement and death in hospitalized cancer patients with severe cutaneous adverse reactions. Journal of the American Academy of Dermatology 2019;80:608–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Correia O, Delgado L, Barbosa IL, Campilho F , Fleming-Torrinha J. Increased interleukin 10, tumor necrosis factor α, and interleukin 6 levels in blister fluid of toxic epidermal necrolysis. Journal of the American Academy of Dermatology 2002;47:58–62. [DOI] [PubMed] [Google Scholar]
- 8.Wang F, He D, Tang X , Zhang X. Chemokine expression in diverse nonimmediate drug hypersensitivity reactions: focus on thymus activation-regulated chemokine, cutaneous T-cell–attracting chemokine, and interleukin-10. Annals of Allergy, Asthma & Immunology 2014;113:204–8. [DOI] [PubMed] [Google Scholar]
- 9.Kardaun SH, Sidoroff A, Valeyrie-Allanore L, Halevy S, Davidovici BB, Mockenhaupt M et al. Variability in the clinical pattern of cutaneous side-effects of drugs with systemic symptoms: does a DRESS syndrome really exist? The British journal of dermatology 2007;156:609–11. [DOI] [PubMed] [Google Scholar]
- 10.Greco R, Lorentino F, Nitti R, Lupo Stanghellini MT, Giglio F, Clerici D et al. Interleukin-6 as Biomarker for Acute GvHD and Survival After Allogeneic Transplant With Post-transplant Cyclophosphamide. Front Immunol 2019;10:2319-. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Shiohara T, Mizukawa Y , Aoyama Y. Monitoring the acute response in severe hypersensitivity reactions to drugs. Current opinion in allergy and clinical immunology 2015;15:294–9. [DOI] [PubMed] [Google Scholar]
- 12.Le RQ, Li L, Yuan W, Shord SS, Nie L, Habtemariam BA et al. FDA Approval Summary: Tocilizumab for Treatment of Chimeric Antigen Receptor T Cell-Induced Severe or Life-Threatening Cytokine Release Syndrome. Oncologist 2018;23:943–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Kochenderfer JN, Dudley ME, Feldman SA, Wilson WH, Spaner DE, Maric I et al. B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. Blood 2012;119:2709–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature 2006;441:235–8. [DOI] [PubMed] [Google Scholar]
- 15.Veldhoen M, Hocking RJ, Atkins CJ, Locksley RM , Stockinger B. TGFβ in the Context of an Inflammatory Cytokine Milieu Supports De Novo Differentiation of IL-17-Producing T Cells. Immunity 2006;24:179–89. [DOI] [PubMed] [Google Scholar]
- 16.Yu C, Chang C , Zhang J. Immunologic and genetic considerations of cutaneous lupus erythematosus: a comprehensive review. Journal of autoimmunity 2013;41:34–45. [DOI] [PubMed] [Google Scholar]
- 17.Chen X, Das R, Komorowski R, Beres A, Hessner MJ, Mihara M et al. Blockade of interleukin-6 signaling augments regulatory T-cell reconstitution and attenuates the severity of graft-versus-host disease. Blood 2009;114:891–900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Feghali CA, Bost KL, Boulware DW , Levy LS. Mechanisms of pathogenesis in scleroderma. I. Overproduction of interleukin 6 by fibroblasts cultured from affected skin sites of patients with scleroderma. The Journal of rheumatology 1992;19:1207–11. [PubMed] [Google Scholar]
- 19.Grossman RM, Krueger J, Yourish D, Granelli-Piperno A, Murphy DP, May LT et al. Interleukin 6 is expressed in high levels in psoriatic skin and stimulates proliferation of cultured human keratinocytes. Proceedings of the National Academy of Sciences of the United States of America 1989;86:6367–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Lee CE, Neuland ME, Teaford HG, Villacis BF, Dixon PS, Valtier S et al. Interleukin-6 is released in the cutaneous response to allergen challenge in atopic individuals. The Journal of allergy and clinical immunology 1992;89:1010–20. [DOI] [PubMed] [Google Scholar]
- 21.Fleming TE, Mirando WS, Soohoo LF, Cooper BW, Zaim MT, Lazarus HM et al. An inflammatory eruption associated with recombinant human IL-6. The British journal of dermatology 1994;130:534–6. [DOI] [PubMed] [Google Scholar]
- 22.Johnson DE, O’Keefe RA , Grandis JR. Targeting the IL-6/JAK/STAT3 signalling axis in cancer. Nature reviews Clinical oncology 2018;15:234–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Kumari N, Dwarakanath BS, Das A , Bhatt AN. Role of interleukin-6 in cancer progression and therapeutic resistance. Tumour biology : the journal of the International Society for Oncodevelopmental Biology and Medicine 2016;37:11553–72. [DOI] [PubMed] [Google Scholar]
- 24.Zhang L, Yang J, Qian J, Li H, Romaguera JE, Kwak LW et al. Role of the microenvironment in mantle cell lymphoma: IL-6 is an important survival factor for the tumor cells. Blood 2012;120:3783–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Bialkowski L, Van der Jeught K, Bevers S, Tjok Joe P, Renmans D, Heirman C et al. Immune checkpoint blockade combined with IL-6 and TGF-beta inhibition improves the therapeutic outcome of mRNA-based immunotherapy. International journal of cancer 2018;143:686–98. [DOI] [PubMed] [Google Scholar]
- 26.Li J, Xu J, Yan X, Jin K, Li W , Zhang R. Targeting Interleukin-6 (IL-6) Sensitizes Anti-PD-L1 Treatment in a Colorectal Cancer Preclinical Model. Medical science monitor : international medical journal of experimental and clinical research 2018;24:5501–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Drobyski WR, Pasquini M, Kovatovic K, Palmer J, Douglas Rizzo J, Saad A et al. Tocilizumab for the treatment of steroid refractory graft-versus-host disease. Biol Blood Marrow Transplant 2011;17:1862–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Kennedy GA, Varelias A, Vuckovic S, Le Texier L, Gartlan KH, Zhang P et al. Addition of interleukin-6 inhibition with tocilizumab to standard graft-versus-host disease prophylaxis after allogeneic stem-cell transplantation: a phase 1/2 trial. The Lancet Oncology 2014;15:1451–9. [DOI] [PubMed] [Google Scholar]
- 29.Roddy JV, Haverkos BM, McBride A, Leininger KM, Jaglowski S, Penza S et al. Tocilizumab for steroid refractory acute graft-versus-host disease. Leukemia & lymphoma 2016;57:81–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Givon T, Revel M , Slavin S. Potential use of interleukin-6 in bone marrow transplantation: effects of recombinant human interleukin-6 after syngeneic and semiallogeneic bone marrow transplantation in mice. Blood 1994;83:1690–7. [PubMed] [Google Scholar]
- 31.Viard-Leveugle I, Gaide O, Jankovic D, Feldmeyer L, Kerl K, Pickard C et al. TNF-alpha and IFN-gamma are potential inducers of Fas-mediated keratinocyte apoptosis through activation of inducible nitric oxide synthase in toxic epidermal necrolysis. The Journal of investigative dermatology 2013;133:489–98. [DOI] [PubMed] [Google Scholar]
- 32.Gencoglan G, Tosun M , Aktepe F. The molecular mechanism of etanercept, an anti-tumour necrosis factor-a receptor-fusion protein, in the treatment of acute generalized exanthematous pustulosis. Journal of Dermatological Treatment 2009;20:241–5. [DOI] [PubMed] [Google Scholar]
- 33.Levine JE. Implications of TNF-α in the pathogenesis and management of GVHD. Int J Hematol 2011;93:571–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Posadas SJ, Padial A, Torres MJ, Mayorga C, Leyva L, Sanchez E et al. Delayed reactions to drugs show levels of perforin, granzyme B, and Fas-L to be related to disease severity. The Journal of allergy and clinical immunology 2002;109:155–61. [DOI] [PubMed] [Google Scholar]
- 35.Yang Y, Li F, Du J, Shen Y, Lin J, Zhu X et al. Variable levels of apoptotic signal-associated cytokines in the disease course of patients with Stevens-Johnson syndrome and toxic epidermal necrolysis. The Australasian journal of dermatology 2017;58:e61–e7. [DOI] [PubMed] [Google Scholar]
- 36.Balkwill F TNF-alpha in promotion and progression of cancer. Cancer metastasis reviews 2006;25:409–16. [DOI] [PubMed] [Google Scholar]
- 37.Wu Y , Zhou BP. TNF-alpha/NF-kappaB/Snail pathway in cancer cell migration and invasion. British journal of cancer 2010;102:639–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Michalaki V, Syrigos K, Charles P , Waxman J. Serum levels of IL-6 and TNF-α correlate with clinicopathological features and patient survival in patients with prostate cancer. British journal of cancer 2004;90:2312–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Harrison ML, Obermueller E, Maisey NR, Hoare S, Edmonds K, Li NF et al. Tumor necrosis factor alpha as a new target for renal cell carcinoma: two sequential phase II trials of infliximab at standard and high dose. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 2007;25:4542–9. [DOI] [PubMed] [Google Scholar]
- 40.Wang H, Liu J, Hu X, Liu S , He B. Prognostic and Therapeutic Values of Tumor Necrosis Factor-Alpha in Hepatocellular Carcinoma. Medical science monitor : international medical journal of experimental and clinical research 2016;22:3694–704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Leman RE, Chen L, Shi X, Rolimpandoei SP, Ling X , Su Y. Drug reaction with eosinophilia and systemic symptoms (DRESS) successfully treated with tumor necrosis factor-α inhibitor. JAAD Case Reports 2017;3:332–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Paradisi A, Abeni D, Bergamo F, Ricci F, Didona D , Didona B. Etanercept therapy for toxic epidermal necrolysis. Journal of the American Academy of Dermatology 2014;71:278–83. [DOI] [PubMed] [Google Scholar]
- 43.Zhang S, Tang S, Li S, Pan Y , Ding Y. Biologic TNF-alpha inhibitors in the treatment of Stevens-Johnson syndrome and toxic epidermal necrolysis: a systemic review. The Journal of dermatological treatment 2019:1–8. [DOI] [PubMed] [Google Scholar]
- 44.Wang CW, Yang LY, Chen CB, Ho HC, Hung SI, Yang CH et al. Randomized, controlled trial of TNF-alpha antagonist in CTL-mediated severe cutaneous adverse reactions. The Journal of clinical investigation 2018;128:985–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Coondoo A Cytokines in dermatology - a basic overview. Indian J Dermatol 2011;56:368–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Nassif A, Moslehi H, Le Gouvello S, Bagot M, Lyonnet L, Michel L et al. Evaluation of the Potential Role of Cytokines in Toxic Epidermal Necrolysis. Journal of Investigative Dermatology 2004;123:850–5. [DOI] [PubMed] [Google Scholar]
- 47.Enk AH , Katz SI. Identification and induction of keratinocyte-derived IL-10. The Journal of Immunology 1992;149:92. [PubMed] [Google Scholar]
- 48.Boulland ML, Meignin V, Leroy-Viard K, Copie-Bergman C, Brière J, Touitou R et al. Human interleukin-10 expression in T/natural killer-cell lymphomas: Association with anaplastic large cell lymphomas and nasal natural killer- cell lymphomas. American Journal of Pathology 1998;153:1229–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Boyano MD, Garcia-Vázquez MD, López-Michelena T, Gardeazabal J, Bilbao J, Canavate ML et al. Soluble interleukin-2 receptor, intercellular adhesion molecule-1 and interleukin-10 serum levels in patients with melanoma. British journal of cancer 2000;83:847–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Li C, Li H, Jiang K, Li J , Gai X. TLR4 signaling pathway in mouse Lewis lung cancer cells promotes the expression of TGF-β1 and IL-10 and tumor cells migration. Bio-Medical Materials and Engineering 2014;24:869–75. [DOI] [PubMed] [Google Scholar]
- 51.Giovarelli M, Musiani P, Modesti A, Dellabona P, Casorati G, Allione A et al. Local release of IL-10 by transfected mouse mammary adenocarcinoma cells does not suppress but enhances antitumor reaction and elicits a strong cytotoxic lymphocyte and antibody-dependent immune memory. Journal of Immunology 1995;155:3112–23. [PubMed] [Google Scholar]
- 52.Zhao S, Wu D, Wu P, Wang Z , Huang J. Serum IL-10 Predicts Worse Outcome in Cancer Patients: A Meta-Analysis. PLoS One 2015;10:e0139598–e. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Schindler K, Harmankaya K, Kuk D, Mangana J, Michielin O, Hoeller C et al. Correlation of absolute and relative eosinophil counts with immune-related adverse events in melanoma patients treated with ipilimumab. Journal of Clinical Oncology 2014;32. [Google Scholar]