Next Article in Journal
Sequential and Hybrid PET/MRI Acquisition in Follow-Up Examination of Glioblastoma Show Similar Diagnostic Performance
Next Article in Special Issue
Immune Checkpoint Inhibitors for Solid Tumors in the Adjuvant Setting: Current Progress, Future Directions, and Role in Transplant Oncology
Previous Article in Journal
Exosome-Derived microRNA: Implications in Melanoma Progression, Diagnosis and Treatment
Previous Article in Special Issue
Recent Trends and Advancements in the Diagnosis and Management of Gastric Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Impact of Infections in Patients Receiving Pembrolizumab-Based Therapies for Non-Small Cell Lung Cancer

1
Houston Methodist Neal Cancer Center, Houston Methodist Hospital, 6445 Main St. Outpatient Center, Floor 24, Houston, TX 77030, USA
2
Department of Medicine, Houston Methodist Hospital, 6565 Fannin St., Smith Tower, Floor 10, Houston, TX 77030, USA
3
Center for Health Data Science and Analytics, Houston Methodist Research Institute, 7550 Greenbriar RB4-129, Houston, TX 77030, USA
4
Section of Hematology and Oncology, Department of Medicine, Baylor College of Medicine, 7200 Cambridge St. 7th Fl, Houston, TX 77030, USA
5
Section of Hematology and Oncology, Scripps Health, 10666 N. Torrey Pines Rd., La Jolla, CA 92037, USA
6
Department of Infectious Diseases, Houston Methodist Hospital, 6550 Fannin St., Ste 1101, Houston, TX 77030, USA
7
Department of Pharmacy, Houston Methodist Hospital, 6565 Fannin St, Houston, TX 77030, USA
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(1), 81; https://doi.org/10.3390/cancers15010081
Submission received: 4 December 2022 / Revised: 15 December 2022 / Accepted: 20 December 2022 / Published: 23 December 2022
(This article belongs to the Special Issue Immunotherapy and Transplantation in the Era of Transplant Oncology)

Abstract

:

Simple Summary

Infections are reported in up to 19% of pembrolizumab treated patients across clinical trials and in 7–100% in the standard of care population receiving various immune checkpoint inhibitor therapies. Risk factors for infection development remains controversial but among them include immunosuppression for the management of immune related adverse events. This study assessed the incidence, type, and impact of infection on morbidity and mortality in non-small cell lung cancer patients receiving pembrolizumab-based therapies. Infections occurred in 46% patients, resulted in a 70% hospitalization rate, treatment delays in 50%, and poorer median overall survival. Furthermore, poorer performance status and anti-infective use at therapy initiation were associated with a higher risk of developing an infection. Infections should be uniformly reported across clinical trials. Anti-infective stewardship may be beneficial to augment treatment adherence and efficacy.

Abstract

Background: Immune checkpoint inhibitor (ICI) therapy has significantly improved outcomes across a range of malignancies. While infections are a well-known contributor to morbidity and mortality amongst patients receiving systemic chemotherapy regimens, little is known about the impact of infections on patients receiving ICI therapy. This study aims to assess incidence, risk factors, and outcomes in patients who develop infections while on pembrolizumab-based therapies for non-small cell lung cancer (NSCLC). Methods: Patients receiving pembrolizumab for stage III/IV NSCLC from 1/1/2017-8/1/2021 across seven hospitals were identified. Incidence and type of infection were characterized. Covariates including baseline demographics, treatment information, treatment toxicities, and immunosuppressive use were collected and compared between infected and non-infected patients. Outcomes included the rate of infections, all-cause hospital admissions, median number of treatment cycles, overall survival (OS), and progression free survival (PFS). Univariable and multivariable analysis with reported odds ratio (OR) and 95% confidence intervals (CI) were utilized to evaluate infection risks. OS and PFS were analyzed by Kaplan–Meier analysis and tested by log-rank test. p-value < 0.05 was considered statistically significant. Results: There were 243 NSCLC patients that met the inclusion criteria. Of these, 111 (45.7%) had one documented infection, and 36 (14.8%) had two or more. Compared to non-infected patients, infected patients had significantly more all-cause Emergency Department (ED) [37 (33.3%) vs. 26 (19.7%), p = 0.016], hospital [87 (78.4%) vs. 53 (40.1%), p < 0.001], and ICU visits [26 (23.4%) vs. 5 (3.8%), p < 0.001], and had poorer median OS (11.53 [95% CI 6.4–16.7] vs. 21.03 [95% CI: 14.7–24.2] months, p = 0.033). On multivariable analysis, anti-infective therapy (OR 3.32, [95% CI: 1.26–8.76], p = 0.015) and ECOG of >1 (OR 5.79, [95% CI 1.72–19.47], p = 0.005) at ICI initiation conferred an increased risk for infections. At last evaluation, 74 (66.7%) infected and 70 (53.0%) non-infected patients died (p = 0.041). Conclusion: Infections occurred in nearly half of patients receiving pembrolizumab-based therapies for NSCLC. Infected patients had frequent hospitalizations, treatment delays, and poorer survival. ECOG status and anti-infective use at ICI initiation conferred a higher infection risk. Infection prevention and control strategies are needed to ameliorate the risk for infections in patients receiving ICIs.

1. Background

Immune checkpoint inhibitor (ICI) therapy has changed the therapeutic framework and improved outcomes across a range of malignancies in the neoadjuvant, adjuvant, and metastatic settings. In non-squamous NSCLC, long term follow-up of the pivotal Keynote 021 study found that the combination of carboplatin and pemetrexed with pembrolizumab yielded superior OS and PFS versus chemotherapy alone, regardless of programmed death ligand-1 (PDL-1) expression [1]. In the Keynote-407 study assessing outcomes of carboplatin and either paclitaxel or nanoparticle albumin-bound [nab]-paclitaxel with or without pembrolizumab in squamous NSCLC, the patients randomized to the pembrolizumab arm had significantly longer OS and PFS compared to patients receiving chemotherapy alone [2]. Furthermore, pembrolizumab has also been established and approved as a monotherapy following the results reported in the Keynote-024 study which demonstrated superior OS and PFS compared to platinum-based chemotherapy in the cohort with a PDL-1 status ≥50% and has also shown beneficial outcomes as monotherapy with TPS > 1% in the elderly population [3,4].
Immune related adverse events (irAE) are a well-known complication of therapy and occur in up to 25–30% of NSCLC patients [1,2,3,4]. IrAE can impact virtually any organ and are fatal in 0.8–1.3% of patients [5]. While these toxicities are well described, the full gambit of toxicities in this growing treatment population needs to be further explored to maximize therapeutic adherence. Infections, a common cause of morbidity and mortality in patients with solid organ malignancies, are infrequently reported in clinical trials of ICI therapy. Furthermore, while several recent retrospective studies have assessed the incidence of infections in ICI-treated patients, the rate varied dramatically, ranging from 7.4–100% [6,7,8,9,10]. While these studies do not imply causality, they suggest that infections may not be infrequent while on ICI therapy. However, questions remain, including the rate and burden of infections with specific ICIs and malignancies, as well as the effect these infections have on treatment adherence, morbidity and mortality. The aim of this study is to assess the rate and outcomes of infections that occur in NSCLC patients receiving pembrolizumab-based therapies.

2. Materials/Methods

2.1. Study Design

This is an institutional review board (IRB) approved, observational, retrospective study assessing the rate, risk factors, and outcomes of infections in patients receiving pembrolizumab-based systemic therapy regimens for the treatment of advanced or metastatic NSCLC. Electronic charts of individuals who received pembrolizumab between the dates of 1 January 2017–1 August 2021 across 7 academic and community hospitals in a single hospital network were reviewed. Patients were included if they were ≥18 years of age at the time of diagnosis, pathologic diagnosis was available, had a confirmed receipt of ≥1 cycle of pembrolizumab either as monotherapy or in combination with a systemic chemotherapy regimen at a study institution, and if patients had stage III or IV disease at the time of ICI initiation. Patients were excluded if they had missing information including confirmed diagnosis, available treatment records, therapy at an institution outside of the hospitals approved for this study, or stage I/II disease.

2.2. Data Collection and Outcomes

All patient data was individually reviewed for study inclusion. A patient was considered to have an infection if there was relevant microbiological confirmation, or if the patient had clinical findings including symptoms correlated with laboratory evidence or imaging characteristics suspicious for infection. Patients with ambiguous infection status were independently reviewed by an infectious disease specialist. If the patient developed relevant symptoms indicative of infection documented in the outpatient setting, they were included. If no infection was documented anywhere in chart review, then they were considered to not have an infection.
Patients were grouped into the infection cohort if they had an infection while on active treatment with pembrolizumab or within 3 months following discontinuation of ICI therapy and not actively receiving other therapy at the time the infection was diagnosed. Infections were divided into bacterial, viral, fungal, parasitic, and systematically categorized into intracranial, intraabdominal, pulmonary, skin and soft tissue infection (SSTI), urinary tract infection (UTI), head and neck, bacteremia, cardiac, neutropenic fever, COVID-19, and presumed sepsis. If a specific pathogen was isolated, it was recorded and catalogued based on infectious site. The course of each infection, including whether the infection resulted or contributed to an emergency department (ED), inpatient, or intermediate unit (IMU)/intensive care unit (ICU) stay was reported. In addition, the impact on treatment delays, interruptions, discontinuation, or death were recorded. If an infection was suspected to be the direct cause of death, it was assessed by an infectious disease specialist for corroboration. Any uncertainties in treatment management were reviewed by a licensed pharmacist in the Cancer Center.
Additional covariates were collected and compared between infected and noninfected patients, which included age, sex (male/female), baseline medical comorbidities, Eastern Cooperative Oncology Group (ECOG) performance status (ranging from 0 defined as fully active without restrictions to 4, which is completely disabled), race (self-reported), chronic infections (including history of Hepatitis B virus [HBV], Hepatitis C virus [HCV], Human Immunodeficiency Virus [HIV], genital or cutaneous herpes simplex virus infections, tuberculosis, cytomegalovirus, and varicella zoster reactivation), line of therapy in which pembrolizumab was administered (1L, 2L, >2L), monotherapy/combination therapy, anti-infective use at ICI initiation, and growth factor use.
The primary objective of this study was to determine the rate of infections in patients receiving ICI therapy for NSCLC. Secondary objectives included: categorical type of infection; the impact of infections on cancer-related treatment and outcomes; impact on morbidity (defined as all-cause emergency department, inpatient hospitalizations, or IMU/ICU visits); infection-related death; impact on overall survival (OS)/progression free survival (PFS).

3. Statistical Analysis and Outcomes

Baseline characteristics were reported as means ± SD for continuous variables and frequencies and percentages for categorical variables by infection status (infected vs. non-infected). Time to infection was reported as median (range: minimum to maximum) in days. Continuous variables were compared by Student’s t-test or Mann–Whitney test, and categorical variables were compared by Chi-square test or Fisher’s exact test. Univariable and multivariable logistic regression models were used to establish characteristics that were independently associated with infectious risk during ICI therapy. These outcomes were reported as odds ratio (OR), along with 95% confidence intervals (CI) and p-values. Kaplan–Meier analysis was performed between infected or non-infected patients, and log-rank test was used to test the OS and PFS between the two cohorts. Cox proportional hazard ratios reported with associated hazard ratio (HR) and 95% CI were used to assess factors impacting OS and PFS with univariable and multivariable models. All analyses were performed with STATA version 17 (StataCorp. 2021. Stata Statistical Software: Release 17. StataCorp LLC, College Station, TX, USA). Statistical significance was defined as two-tailed p < 0.05 for all tests.

4. Results

4.1. Baseline Factors

Between 1 January 2017 and 1 August 2021, there were 810 evaluable patients, of which 243 patients had NSCLC and met inclusion criteria for analysis (Figure 1). Of these, 111 (45.7%) had at least one infection. Among those infected, there were 93 (83.8%) with adenocarcinoma, 15 (13.5%) with squamous cell carcinoma, and 3 (2.7%) with “other” histology. The mean age of infected patients was 69.12 ± 11 years, 59 (53.15%) were female, 83 (74.77%) identified themselves as white, and the most common pre-existing medical conditions included chronic obstructive pulmonary disease (COPD) in 49 (44.1%) and type 2 diabetes mellitus (T2DM) in 26 (23.42%). Most infected patients had an ECOG of 1 or 2 in 47 (42.34%) and 23 (20.72%), respectively. Out of the 243 patients, 22 (9.0%) had one or more chronic infections at ICI initiation, 15 (13.5%) of which went on to have an infection. Of these 15, 2 (13.3%) had HBV, 4 (26.7%) had HCV, 1 (6.7%) had HIV, 3 (20.0%) had a history of tuberculosis, 7 (46.7%) had HSV, and 1 (6.7%) had HIV. Baseline PD-L1 activity was evaluable in 95 (85.6%) patients with infection; 39 (35.14%) had absent membranous staining, 19 (17.1%) had staining ranging from 1–20%, and 37 (33.33%) had >20% PD-L1 activity.
There were 41 (36.94%) infected patients compared to 39 (29.55%) noninfected on monotherapy, and 70 (63.06%) and 93 (70.45%) on multiagent systemic therapy, respecively. Over 70% of patients in both cohorts were using pembrolizumab-based therapies in 1L. A total of 27 (24.3%) and 38 (28.8%) in infected and non-infected cohorts, respectively required radiation while on an ICI over the course of their disease. At initiation of ICI therapy, 31 (12.7%) patients were on steroids, and 15 (48.4%) of these patients developed infections. Moreover, 21 (18.92%) infected patients were on anti-infectives at the time of ICI initiation (Table 1).

4.2. Infections

Of the 243 patients included, 111 (45.7%) had one infection, 55 (22.6%) had two infections, and 18 (7.4%) had three or more infections (Table 2, Figure 2 and Figure S1). Median time to first, second, and third infection was 58 (1–1465), 124 (6–844), and 213.5 (25–1414) days from initiation of therapy, respectively. IMU/ICU admission as a result of complications arising from infection was required in 13 (11.7%) with one infection, 14 (25.4%) with 2 infections, and 6 (33.3%) patients with ≥3 infections. An infectious organism was confirmed in 53 (47.7%) with one infection, 22 (40.0%) with two infections, and 6 (33.3%) with ≥3 infections; a summary of these organisms can be found in Table S1 of the Supplementary Appendix. Of note, Escherichia coli spp. and Clostridioides difficile were the most commonly isolated bacteria occurring in 11 (9.9%) and 5 (4.5%) patients, while Rhinovirus/Enterovirus, varicella zoster reactivation, and Herpes simplex 1/2 orogenital infections were the most commonly isolated viral infections, occurring in 4 (3.6%), 3 (2.7%), and 3 (2.7%) patients, respectively. The most common sites of infection included pulmonary in 48 (43.2%), urinary tract in 20 (18.0%), and SSTI in 15 (13.5%) patients (Figure S1). COVID-19 pneumonia was diagnosed in 3 (2.7%) patients while receiving pembrolizumab. There were a total of 7 (6.3%) fungal infections isolated, of which 3 (42.8%) were on monotherapy and 4 (57.1%) were on combination therapy. Of the 15 patients with chronic infections, 3 patients with known HSV developed reactivation with orogenital infections; no other reactivation of infections occurred, including in those not on active therapy for their disease at the time (Table S1).
For treatment, 104 patients (93.7%) received antimicrobials, 12 (10.8%) received antiviral therapy, and 10 (9.0%) received antifungal therapy. The most commonly prescribed antibiotic classes were cephalosporins and vancomycin, prescribed 64 (29.6%) and 53 (24.5%) times each, respectively (Table S2). A total of 47 (42.3%) patients had an infectious disease consultation at least once.
On univariable analysis, COPD (p = 0.013), and ECOG performanc status of 1 (p = 0.027), 2 (p = 0.001), or unknown (p = 0.001) ECOG status, history of chronic infection (p = 0.032), and anti-infective at time of ICI initiation (p = 0.003) were associated with higher risk of developing infection. However, on multivariable analysis, only ECOG 2 (OR 5.79 [95% CI 1.72, 19.47], p = 0.005), unknown ECOG (4.80 [95% CI 1.57, 14.74], p = 0.006), and anti-infective use at ICI initiation (OR 3.32 [95% CI 1.26, 8.76], p = 0.015) retained a statistically significant risk of developing an infection while on ICI therapy (Table 3).

4.3. Outcomes

A total of 66 (27.16%) patients developed an irAE event, of which 33 (29.73%) occurred in the infection cohort (p = 0.41) (Table 4). Pneumonitis was more common in the infected cohort on univariable analysis (p = 0.009). Over the course of workup or management of documented infections, treatment delays occurred in 56 (50.4%), treatment discontinuation in 28 (25.2%), and disease progression was documented in 16 (14.4%) patients (Table S2, Figure S1). There were significantly more all-cause ED [37 (33.3%) vs. 26 (19.7%), p = 0.016], hospital [87 (78.38%) vs. 53 (40.15%), p < 0.001], and IMU/ICU visits [26 (23.42%) vs. 5 (3.79%), p < 0.001] in infected compared to non-infected patients, respectively. At the last follow up, there were a total of 144 (59.26%) deaths, of which 74 (66.7%) and 70 (53.03%) occurred in infected and noninfected, respectively (p = 0.031). Of these, 11 (14.86%) with infections died due to complications arising from the infection; 4 (5.71%) in the noninfected group died due to eventual infection but were not included in the infection cohort because they had an infection while on subsequent treatment or had an infection >3 months after ICI discontinuation (Table 4).
Regarding timing of infections, 30/111 (27.0%) developed an infection during the first two cycles of therapy, which comprised 54.5% of the 55 patients who received only two cycles of therapy. Moreover, 6/11 (54.5%) of the patients that died as a direct cause of infection died after receiving one (n = 5) or two (n = 1) cycles. There were 11 patients with infections that had disease progression within the first two cycles, of which seven (63.6%) died from primary disease and one (9.1%) died from infection. There were another 23 that received the first two cycles who did not have disease progression reported; however 10 (43.5%) deaths were due underlying primary disease burden, five (21.7%) were due to infection, and two (8.7%) had multiorgan failure.
Median OS was 11.53 [95% CI 6.37–16.73] months for infected and 21.03 [95% CI 14.7–24.17] months for non-infected (p = 0.033) (Figure 2A). Median OS for patients on monotherapy and no infection was 22.33 (95% CI 11.4–46.53) months compared to monotherapy with infection which was 14.17 months (95% CI: 5.4–24.5), and for patients on multiagent therapy, was 19.73 months (95% CI: 12.03–31.03) without an infection, and 10.33 months (95% CI 5.77–16.53) with an infection (Figure S2). However, when controlling for treatment and infection status, these differences were not statistically significant (p = 0.12). Of note, while 47 (42.3%) of infected patients required an infectious disease consultation, this did not significantly impact survival (Figure S2B). Median PFS in infected and non-infected cohorts was 5.9 (95% CI 4.16, 7.6) and 7.9 (95% CI 6.94, 11.03) (p = 0.066) months, respectively (Figure 2B). On multivariable analysis, age (HR 1.02 [95% CI 1.00, 1.04], p = 0.019), T2DM (HR 1.58 [95% CI 1.07, 2.33], p = 0.022), and ECOG 2 (HR 2.34 [95% CI 1.08, 5.08], p = 0.031), had a higher mortality rate, whereas patients that received 3 or more cycles of treatment (HR 0.33 [95% CI 0.20, 0.53], p < 0.001) had better OS. With regard to PFS, COPD (HR 0.59 [95% CI 0.40, 0.88], p = 0.010) had a better reported PFS, whereas patients on 2L (HR 1.74 [95% CI 1.15, 2.63], p = 0.009) and >2L (HR 2.28 [95% CI 1.20, 4.33], p = 0.011) had higher rate of progression compared to patients receiving pembrolizumab in 1L.

5. Discussion

Infections impart a substantial risk for morbidity and mortality in patients with solid organ malignancies. However, there is scarce data assessing the burden and impact of infections occurring with specific ICI therapies. We recently reported upon outcomes in patients receiving pembrolizumab for head and neck squamous cell carcinomas (HNSCC) and urothelial adenocarcinomas [11,12], indicating that infections increase the number of all-cause hospitalizations and are associated with poorer survival. Herein, we report the rate and impact of infections in patients with NSCLC who received pembrolizumab. The following key findings were found: infections occurred in nearly half of patients, with the most common subtype of infection being bacterial pulmonary infections; anti-infective use and poorer ECOG performance status at pembrolizumab initiation were associated with a higher risk of infection; infected patients had significantly higher rate of all-cause ED, hospital, and IMU/ICU admissions; infection status negatively impacted OS and resulted in death in approximately 10% of patients.
The risk and impact of infection in patients receiving ICI therapy remains controversial. A recent review of trial data by the European Society of Clinical Microbiology and Infectious Diseases (ESCMID) led to the conclusion that that CTLA-4, PD-1, and PD-L1 inhibitors are not independently associated with risk of infection [13]. However, the package insert for pembrolizumab cautions that genitourinary tract infections are reported in up to 19% of patients, upper respiratory tract in 13–44%, and pneumonia in 12–19% of patients [14]. While clinical trials are the gold standard for determining treatment efficacy, they may not adequately assess infrequent toxicities, particularly if causality between the medication and event is not clearly established. Post-marketing studies are helpful in these cases to assess outcomes of less common toxicities in the standard of care population that may be more heterogenous than the trial cohorts. Until recently, few studies have assessed the spectrum, burden, and impact of infections. In 2016, an analysis of infection occurrence in melanoma patients receiving nivolumab or pembrolizumab with or without ipilimumab determined that the rate of “serious” infections requiring hospitalization occurred in 7.3% of cases [8]. In a recent meta-analysis that assessed over 21,000 patients from over 36 studies, ICI monotherapy had similar risks for all-grade infections (relative risk [RR] = 1.02; 95% CI 0.84–1.24; p = 0.85), a lower risk of infection compared to chemotherapy alone (RR = 0.58, 95% CI 0.4–0.85; p = 0.01), and a higher risk than chemotherapy alone when combined with chemotherapy (RR = 1.37, 95% CI 1.23–1.53; p < 0.01) or when dual-ICI therapy was used 2.26 (95% CI 1.34–3.8; p < 0.01) [9]. In a study of 111 patients assessing infections in different ICI therapies for a range of cancers, 24% had a suspected or confirmed bacterial infection, but only one viral infection was reported [7]. In a French chart review, the infection rate was 18%, with most common infections being pneumonia in 58%, SSTI and UTI occurring in 19.4% each [10]. In our study, 46% of NSCLC receiving pembrolizumab developed an infection, resulting in a hospitalization rate of nearly 70%. This suggests a highly heterogenous population across post-marketing studies, likely stemming from age, ECOG, baseline demographic factors, and infection severity. This analysis only assessed the NSCLC population treated with pembrolizumab-based therapies, but our recent reports assessing HNSCC and urothelial adenocarcinomas reported similar rates of infection and hospitalization rates [11,12]. Similar to other studies, pulmonary infections, SSTI, and UTIs were the most common infections, but our analysis found a higher number of viral and fungal infections.
This study was not designed to assess causality between pembrolizumab and infections, but rather to assess the rate and risk of infections in NSCLC patients receiving pembrolizumab-based therapies. Risk factors for developing infection in this study included a poorer ECOG performance status and anti-infective use at pembrolizumab initiation. Similar findings were also seen in patients receiving pembrolizumab for HNSCC [12]. Notably, our study did not find an increased risk for infection in patients receiving concomitant chemotherapy, radiation therapy, or in patients who had immunosuppressive therapy at the initiation of pembrolizumab or who received immunosuppression to manage irAE. Patients with irAE may be at increased risk for infection by several mechanisms, including immune dysregulation stemming from ICI therapy, local tissue damage, or from immunosuppressive therapies used to ameliorate effects of irAE, such as corticosteroids, infliximab, cyclophosphamide, and mycophenolate mofetil [15]. Sutthichai et al. evaluated the incidence of infection in those on ICI therapy in solid tumor and hematologic malignancies and found that 68% of patients were found to have de novo infections while on an ICI, and all of these patients were started on immunosuppression for irAEs. The study suggested that some patients on ICI therapies, whether as a consequence of the ICI or specifically irAEs, will develop infections [6]. Several other recent studies found that immunosuppressive use, particularly systemic corticosteroids, predisposed patients to infections [7,8,9]. Conversely, like the present study, there are other findings that would suggest factors aside from immunosuppressive use may drive the risk of infection development [10,11]. It has been proposed that immunity dysregulation can occur with the excessive immune response from ICIs and that this in of itself can favor development of infections [9]. Moreover, polymorphisms in PD-1, PD-L1, and CTLA-4 have been associated with the development of a diverse array of infections, suggesting a direct correlation between immune dysregulation and infection development [9,16,17,18].
Our study found that anti-infective use at ICI initiation was associated with a higher infection risk. There is a growing body of literature that anti-infective use negatively impacts ICI efficacy. It is feasible that the association between the higher infection rate in our study stems from an iatrogenic alteration of the normal intestinal microbiota. The intestinal microbiome has several vital functions including the maturation of host immune responses, antigenic recognition, providing protection against bacterial overgrowth, modifying specific drugs, and removing toxins [19]. While individual responses to ICI therapy is derived from a combination of immune competency, diversity, and adequate antigenic variation, recognition, and expression, the intestinal microbiota has recently been found to also play a pivotal role, which arises from the development of tumor antigenicity, adjuvanticity, and bystander activation through an enhanced inflammatory state in the tumor micro-environment [19,20,21,22]. Anti-infectives produce gut micriobiota dysbiosis resulting in loss of diversity, alteration in the basal metabolic capactity, and possible introduction of invasive pathogens [23]. While these changes occur within several days of antimicrobial exposure, it may take up to 6 weeks or more for the intestinal flora to recover [24]. These dramatic changes also diminish the response of cytotoxic T-cells to cancer antigens, and data suggests that these patients have inferior responses to anti-PD-1 ICI therapy and have a lower taxonomic intestinal diversity [25,26,27]. A recent meta-analysis demonstrated poorer OS in patients that received antimicrobials in the month preceding (hazard ratio [HR] 1.64, 95% CI 1.27–2.11) ICI or in the month following ICI therapy (HR 2.36, 95% CI 1.66–2.33), as well as poorer PFS (HR 1.52, 95% CI 1.36–1.70), with the latter being more prominent in patients with RCC or melanoma than with NSCLC [20]. Multiple other studies assessing outcomes of antibiotics in NSCLC treated with ICI therapies have shown negative impacts on OS and PFS, primarily in patients receiving an anti-infective in the preceding 6 weeks up to the first 4 weeks of anti-PD-1 ICI initiation [28,29,30,31,32,33,34]. It is possible that the poorer outcomes in the infected group of this study was similarly impacted. However, while PFS was numerically lower in our cohort, it was not significantly different than the non-infection cohort. This may be explained by the higher early mortality rate stemming from multiorgan failure and infections in the infection cohort. With this accumulating data, anti-infective stewardship, a frequently underutilized strategy in healthcare, may be beneficial to minimize unnecessary anti-infective use. Replenishment of the intestinal microbiome may also be a feasible strategy, particularly in the infected population. A recent meta-analysis exploring the clinical impact of probiotics in patients with NSCLC who received ICIs reported a superior OS (HR = 0.50, 95% CI: 0.30–0.85, p = 0.01) and PFS (HR = 0.51, 95% CI: 0.42–0.61, p < 0.01), suggesting this may be a viable strategy [35].
Limitations of this study should be recognized. As a retrospective chart review, there will be biases intrinsic to the study design itself. Similar to the limited number of other studies currently available, our reported number of infections were notably higher indicating heterogeneity across study populations, likely stemming from underlying population-level differences in baseline comorbidities, performance status, and treatment. In addition, several analyses were based on small sample sizes and as such this led to wide CIs and may have impacted the true clinical significance of the outcomes reported. Furthermore, we did not have enough information or a population large enough to perform a subgroup analysis on the class of anti-infective used, duration of utilization, route of administration, or type of chronic infection. Finally, given the retrospective nature of this chart review study, patients were excluded from the analysis if they did not have relevant information including progress notes, imaging studies, or therapy records (n = 20). This exclusion may have impacted the final results of the multivariable analysis as well as survival differences given the current sample size.

6. Conclusions

Infections in patients receiving pembrolizumab-based therapies for NSCLC are common and result in significantly higher rates of all-cause ED, hospital, and IMU/ICU admissions. Furthermore, infections resulted in death in 10% of infected patients and shorter median OS compared to non-infected patients. Risks for infection development included poorer ECOG performance status and anti-infective use at initiation of pembrolizumab. Studies to assess the benefit of anti-infective stewardships and reduction of anti-infective use at initiation of ICI therapy are needed.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cancers15010081/s1, Figure S1: Distribution of infections; Figure S2A: Median overall survival based on monotherapy and chemotherapy in infected and non-infected patients; Figure S2B: Median overall survival based on infectious disease consultation status in infected patients; Table S1: Cultured infectious organisms based on infection site; Table S2: Outcomes of infection, anti-infectives used in treatment.

Author Contributions

E.A.B., R.B.K., I.N.M., S.S., G.U., M.A.: Conceptualization; E.A.B., J.X., I.N.M., S.S., G.U., M.A.: Methodology; E.A.B., J.X., R.B.K., S.S., G.U., M.A.: Validation; E.A.B., J.X.: Formal Analysis; E.A.B., K.G., R.B.K., J.X., Y.Z., A.C., I.N.M., C.M., A.E., S.S., G.U., K.S., C.G., Z.G., K.H., M.S., J.Z., E.H.B., M.A.: Investigation; G.U., S.S.: Resources; E.A.B., K.G., I.N.M., R.B.K., Y.Z., A.C., C.M., A.E., S.S., G.U., K.S., C.G., Z.G.: Data Curation; E.A.B., K.G.: Writing Original Draft; E.A.B., K.G., R.B.K., J.X., Y.Z., A.C., I.N.M., C.M., A.E., S.S., G.U., K.S., C.G., Z.G., K.H., M.S., J.Z., E.H.B., M.A.: Review and Editing; E.A.B., J.X.: Visualization; K.H., M.S., J.Z., E.H.B., M.A.: Supervision; E.A.B., M.A.: Project Administration. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

IThe study was conducted in accordance with the Declaration of Helsinki, and approved by the Methodist Online Research Technology Initiative Institutional Review Board (PRO00028635).

Informed Consent Statement

As this was a minimal risk chart review study, individual patient consent was not obtained and was not required by the IRB.

Data Availability Statement

Data was made available through the charting review system Epic.

Acknowledgments

The authors would like to acknowledge Eva Zsigmond for medical editing of this manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

NSCLCNon-small cell lung cancer
ICIImmune checkpoint inhibitor
irAEImmune related adverse event
EDEmergency Department
IMUIntermediate Care Unit
ICUIntensive Care Unit
OSOverall survival
PFSProgression free survival
OROdds ratio
CIConfidence interval
ECOGEastern Cooperative Oncology Group
PCRPolymerase Chain Reaction
SSTIsoft tissue/skin infection
UTIUrinary Tract Infection
HBVHepatitis B Virus
HCVHepatitis C Virus
HIVHuman Immunodeficiency
HNSCCHead and Neck Squamous Cell Carcinoma
ESCMIDEuropean Society of Clinical Microbiology and Infectious Diseases
RRRelative Risk
PD-1Programmed Death-1

References

  1. Awad, M.M.; Gadgeel, S.M.; Borghaei, H.; Patnaik, A.; Yang, J.C.-H.; Powell, S.F.; Gentzler, R.D.; Martins, R.G.; Stevenson, J.P.; Altan, M.; et al. Long-Term Overall Survival From KEYNOTE-021 Cohort G: Pemetrexed and Carboplatin With or Without Pembrolizumab as First-Line Therapy for Advanced Nonsquamous NSCLC. J. Thorac. Oncol. 2021, 16, 162–168. [Google Scholar] [CrossRef] [PubMed]
  2. Paz-Ares, L.; Luft, A.; Vicente, D.; Tafreshi, A.; Gümüş, M.; Mazières, J.; Hermes, B.; Çay Şenler, F.; Csőszi, T.; Fülöp, A.; et al. Pembrolizumab plus Chemotherapy for Squamous Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 379, 2040–2051. [Google Scholar] [CrossRef] [PubMed]
  3. Nosaki, K.; Saka, H.; Hosomi, Y.; Baas, P.; de Castro, G., Jr.; Reck, M.; Wu, Y.L.; Brahmer, J.R.; Felip, E.; Sawada, T.; et al. Safety and efficacy of pembrolizumab monotherapy in elderly patients with PD-L1-positive advanced non-small-cell lung cancer: Pooled analysis from the KEYNOTE-010, KEYNOTE-024, and KEYNOTE-042 studies. Lung Cancer 2019, 135, 188–195. [Google Scholar] [CrossRef] [PubMed]
  4. Reck, M.; Rodríguez–Abreu, D.; Robinson, A.G.; Hui, R.; Csőszi, T.; Fülöp, A.; Gottfried, M.; Peled, N.; Tafreshi, A.; Cuffe, S.; et al. Updated Analysis of KEYNOTE-024: Pembrolizumab Versus Platinum-Based Chemotherapy for Advanced Non-Small-Cell Lung Cancer With PD-L1 Tumor Proportion Score of 50% or Greater. J. Clin. Oncol. 2019, 37, 537–546. [Google Scholar] [CrossRef]
  5. Martins, F.; Sofiya, L.; Sykiotis, G.P.; Lamine, F.; Maillard, M.; Fraga, M.; Shabafrouz, K.; Ribi, C.; Cairoli, A.; Guex-Crosier, Y.; et al. Adverse effects of immune-checkpoint inhibitors: Epidemiology, management and surveillance. Nat. Rev. Clin. Oncol. 2019, 16, 563–580. [Google Scholar] [CrossRef]
  6. Sae-Tia, S.; Naidoo, J.; Mehta, S. Infections in patients receiving immune checkpoint inhibitors. J. Clin. Oncol. 2019, 37 (Suppl. 8), 155. [Google Scholar] [CrossRef]
  7. Ross, J.A.; Komoda, K.; Pal, S.; Dickter, J.; Salgia, R.; Dadwal, S. Infectious complications of immune checkpoint inhibitors in solid organ malignancies. Cancer Med. 2022, 11, 21–27. [Google Scholar] [CrossRef]
  8. del Castillo, M.; Romero, F.A.; Argüello, E.; Kyi, C.; Postow, M.A.; Redelman-Sidi, G. The Spectrum of Serious Infections Among Patients Receiving Immune Checkpoint Blockade for the Treatment of Melanoma. Clin. Infect. Dis. 2016, 63, 1490–1493. [Google Scholar] [CrossRef] [Green Version]
  9. Petrelli, F.; Morelli, A.M.; Luciani, A.; Ghidini, A.; Solinas, C. Risk of Infection with Immune Checkpoint Inhibitors: A Systematic Review and Meta-analysis. Target Oncol. 2021, 16, 553–568. [Google Scholar] [CrossRef]
  10. Karam, J.D.; Noel, N.; Voisin, A.L.; Lanoy, E.; Michot, J.M.; Lambotte, O. Infectious complications in patients treated with immune checkpoint inhibitors. Eur. J. Cancer 2020, 141, 137–142. [Google Scholar] [CrossRef]
  11. Kieser, R.B.; Xu, J.; Burns, E.; Muhsen, I.; Shah, S.M.; Umoru, G.; Mylavarapu, C.; Sun, K.; Zhang, Y.; Crenshaw, A.; et al. Outcomes of patients with advanced urothelial cancer who develop infection while on treatment with pembrolizumab. J. Clin. Oncol. 2022, 40 (Suppl. 16), 4573. [Google Scholar] [CrossRef]
  12. Zhang, Y.; Xu, J.; Muhsen, I.; Burns, E.; Shah, S.M.; Umoru, G.; Mylavarapu, C.; Sun, K.; Crenshaw, A.; Esmail, A.; et al. Infections and their impact on patients on pembrolizumab-based therapies for head and neck cancer. J. Clin. Oncol. 2022, 40, 6035. [Google Scholar]
  13. Redelman-Sidi, G.; Michielin, O.; Cervera, C.; Ribi, C.; Aguado, J.M.; Fernández-Ruiz, M.; Manuel, O. ESCMID Study Group for Infections in Compromised Hosts (ESGICH) Consensus Document on the safety of targeted and biological therapies: An infectious diseases perspective (Immune checkpoint inhibitors, cell adhesion inhibitors, sphingosine-1-phosphate receptor modulators and proteasome inhibitors). Clin. Microbiol. Infect. 2018, 24 (Suppl. 2), S95–S107. [Google Scholar] [PubMed] [Green Version]
  14. Keytruda [Package Insert]. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/125514s110lbl.pdf (accessed on 27 October 2022).
  15. Picchi, H.; Mateus, C.; Chouaid, C.; Besse, B.; Marabelle, A.; Michot, J.-M.; Champiat, S.; Voisin, A.; Lambotte, O. Infectious complications associated with the use of immune checkpoint inhibitors in oncology: Reactivation of tuberculosis after anti PD-1 treatment. Clin. Microbiol. Infect. 2018, 24, 216–218. [Google Scholar] [CrossRef] [Green Version]
  16. Thye, T.; Scarisbrick, G.; Browne, E.N.L.; Chinbuah, M.A.; Gyapong, J.; Osei, I.; Owusu-Dabo, E.; Niemann, S.; Rüsch-Gerdes, S.; Meyer, C.G.; et al. CTLA4 autoimmunity-associated genotype contributes to severe pulmonary tuberculosis in an African population. PLoS ONE 2009, 4, e6307. [Google Scholar] [CrossRef] [Green Version]
  17. Gámez-Díaz, L.; August, D.; Stepensky, P.; Revel-Vilk, S.; Seidel, M.G.; Noriko, M.; Morio, T.; Worth, A.J.; Blessing, J.; Van de Veerdonk, F.; et al. The extended phenotype of LPS-responsive beige-like anchor protein (LRBA) deficiency. J. Allergy Clin. Immunol. 2016, 137, 223–230. [Google Scholar] [CrossRef] [Green Version]
  18. Lo, B.; Fritz, J.M.; Su, H.C.; Uzel, G.; Jordan, M.B.; Lenardo, M.J. CHAI and LATAIE: New genetic diseases of CTLA-4 checkpoint insufficiency. Blood 2016, 128, 1037–1042. [Google Scholar] [CrossRef] [Green Version]
  19. Lynch, S.V.; Pedersen, O. The Human Intestinal Microbiome in Health and Disease. N. Engl. J. Med. 2016, 375, 2369–2379. [Google Scholar] [CrossRef] [Green Version]
  20. Botticelli, A.; Zizzari, I.; Mazzuca, F.; Ascierto, P.A.; Putignani, L.; Marchetti, L.; Napoletano, C.; Nuti, M.; Marchetti, P. Cross-talk between microbiota and immune fitness to steer and control response to anti PD-1/PDL-1 treatment. Oncotarget 2017, 8, 8890–8899. [Google Scholar] [CrossRef] [Green Version]
  21. Tsikala-Vafea, M.; Belani, N.; Vieira, K.; Khan, H.; Farmakiotis, D. Use of antibiotics is associated with worse clinical outcomes in patients with cancer treated with immune checkpoint inhibitors: A systematic review and meta-analysis. Int. J. Infect. Dis. 2021, 106, 142–154. [Google Scholar] [CrossRef]
  22. Fessler, J.; Matson, V.; Gajewski, T.F. Exploring the emerging role of the microbiome in cancer immunotherapy. J. Immunother. Cancer 2019, 7, 108. [Google Scholar] [CrossRef] [PubMed]
  23. Lange, K.; Buerger, M.; Stallmach, A.; Bruns, T. Effects of Antibiotics on Gut Microbiota. Dig. Dis. 2016, 34, 260–268. [Google Scholar] [CrossRef] [PubMed]
  24. Tinsley, N.; Zhou, C.; Tan, G.; Rack, S.; Lorigan, P.; Blackhall, F.; Krebs, M.; Carter, L.; Thistlethwaite, F.; Graham, D.; et al. Cumulative Antibiotic Use Significantly Decreases Efficacy of Checkpoint Inhibitors in Patients with Advanced Cancer. Oncologist 2020, 25, 55–63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Pinato, D.J.; Howlett, S.; Ottaviani, D.; Urus, H.; Patel, A.; Mineo, T.; Brock, C.; Power, D.; Hatcher, O.; Falconer, A.; et al. Association of Prior Antibiotic Treatment With Survival and Response to Immune Checkpoint Inhibitor Therapy in Patients with Cancer. JAMA Oncol. 2019, 5, 1774–1778. [Google Scholar] [CrossRef]
  26. Patel, P.; Poudel, A.; Kafle, S.; Thapa Magar, M.; Cancarevic, I. Influence of Microbiome and Antibiotics on the Efficacy of Immune Checkpoint Inhibitors. Cureus 2021, 13, e16829. [Google Scholar] [CrossRef]
  27. Zheng, Y.; Wang, T.; Tu, X.; Huang, Y.; Zhang, H.; Tan, D.; Jiang, W.; Cai, S.; Zhao, P.; Song, R.; et al. Gut microbiome affects the response to anti-PD-1 immunotherapy in patients with hepatocellular carcinoma. J. Immunother. Cancer 2019, 7, 193. [Google Scholar] [CrossRef] [Green Version]
  28. Giordan, Q.; Salleron, J.; Vallance, C.; Moriana, C.; Clement-Duchene, C. Impact of Antibiotics and Proton Pump Inhibitors on Efficacy and Tolerance of Anti-PD-1 Immune Checkpoint Inhibitors. Front. Immunol. 2021, 12, 716317. [Google Scholar] [CrossRef]
  29. Schett, A.; Rothschild, S.I.; Curioni-Fontecedro, A.; Krähenbühl, S.; Früh, M.; Schmid, S.; Driessen, C.; Joerger, M. Predictive impact of antibiotics in patients with advanced non small-cell lung cancer receiving immune checkpoint inhibitors: Antibiotics immune checkpoint inhibitors in advanced NSCLC. Cancer Chemother. Pharmacol. 2020, 85, 121–131. [Google Scholar] [CrossRef]
  30. Facchinetti, F.; Mazzaschi, G.; Barbieri, F.; Passiglia, F.; Mazzoni, F.; Berardi, R.; Proto, C.; Cecere, F.L.; Pilotto, S.; Scotti, V.; et al. First-line pembrolizumab in advanced non-small cell lung cancer patients with poor performance status. Eur. J. Cancer 2020, 130, 155–167. [Google Scholar] [CrossRef]
  31. Derosa, L.; Hellmann, M.D.; Spaziano, M.; Halpenny, D.; Fidelle, M.; Rizvi, H.; Long, N.; Plodkowski, A.J.; Arbour, K.C.; Chaft, J.E.; et al. Negative association of antibiotics on clinical activity of immune checkpoint inhibitors in patients with advanced renal cell and non-small-cell lung cancer. Ann. Oncol. 2018, 29, 1437–1444. [Google Scholar] [CrossRef]
  32. Dennehy, C.; Mullally, W.; Goggin, C.; Gleeson, J.P.; Greene, J.P.; Power, D.G.; O’Reilly, S.; Collins, D.C.; O’Mahony, D.; Bambury, R.M. Real world experience with pembrolizumab and nivolumab for treatment of non-small cell lung cancer (NSCLC) in an Irish Regional Cancer Center. J. Clin. Oncol. 2018, 36 (Suppl. 15), e21196. [Google Scholar] [CrossRef]
  33. Bagley, S.J.; Dhopeshwarkar, N.; Narayan, V.; Meropol, N.J.; Mamtani, R.; Boursi, B. Impact of antibiotics (ABX) on overall survival (OS) in patients (pts) with advanced non-small-cell lung cancer (aNSCLC) and melanoma (aMel) treated with first-line immune checkpoint inhibition (ICI). J. Clin. Oncol. 2019, 37 (Suppl. 15), e20643. [Google Scholar] [CrossRef]
  34. Ahmed, J.; Kumar, A.; Parikh, K.; Anwar, A.; Knoll, B.M.; Puccio, C.; Chun, H.; Fanucchi, M.; Lim, S.H. Use of broad-spectrum antibiotics impacts outcome in patients treated with immune checkpoint inhibitors. Oncoimmunology 2018, 7, e1507670. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Wan, L.; Wu, C.; Wu, Q.; Luo, S.; Liu, J.; Xie, X. Impact of probiotics use on clinical outcomes of immune checkpoint inhibitors therapy in cancer patients. Cancer Med. 2022. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Consort diagram. NSCLC: Nonsmall cell lung cancer. Missing data includes patients who may have received treatment at one of the seven hospitals but did not have electronic charting information available including progress notes, labs, or treatment notes. The one patient with a hypersensitivity reaction during cycle 1 did not complete a full infusion, and was not included.
Figure 1. Consort diagram. NSCLC: Nonsmall cell lung cancer. Missing data includes patients who may have received treatment at one of the seven hospitals but did not have electronic charting information available including progress notes, labs, or treatment notes. The one patient with a hypersensitivity reaction during cycle 1 did not complete a full infusion, and was not included.
Cancers 15 00081 g001
Figure 2. (A) Median overall survival. (B) Progression free survival.
Figure 2. (A) Median overall survival. (B) Progression free survival.
Cancers 15 00081 g002
Table 1. Baseline demographics.
Table 1. Baseline demographics.
Infection
YesNoTotalp-Value
N = 111N = 132N = 243
Age69.12 ± 11.0067.68 ± 11.2868.34 ± 11.150.32
Female59 (53.15%)56 (42.42%)115 (47.33%)0.095
Race 0.85
White83 (74.77%)97 (73.48%)180 (74.07%)
Black18 (16.22%)20 (15.15%)38 (15.64%)
Other Comorbidities10 (9.01%)15 (11.36%)25 (10.29%)
T2DM26 (23.42%)34 (25.76%)60 (24.69%)0.67
CAD33 (29.73%)27 (20.45%)60 (24.69%)0.095
ESRD1 (0.90%)0 (0.00%)1 (0.41%)0.27
COPD49 (44.14%)38 (28.79%)87 (35.80%)0.013
IV Drug Use0 (0.00%)1 (0.76%)1 (0.41%)0.36
Alcoholism5 (4.50%)10 (7.58%)15 (6.17%)0.32
Chronic Indwelling Catheter3 (2.70%)1 (0.76%)4 (1.65%)0.24
Smoking Status 0.30
Never13 (11.71%)21 (15.91%)34 (13.99%)
Previous81 (72.97%)84 (63.64%)165 (67.90%)
Current17 (15.32%)27 (20.45%)44 (18.11%)
ECOG 0.001
05 (4.50%)23 (17.42%)28 (11.52%)
147 (42.34%)67 (50.76%)114 (46.91%)
223 (20.72%)13 (9.85%)36 (14.81%)
33 (2.70%)3 (2.27%)6 (2.47%)
40 (0.00%)1 (0.76%)1 (0.41%)
Unknown33 (29.73%)25 (18.94%)58 (23.87%)
Previous ICI therapy6 (5.41%)8 (6.06%)14 (5.76%)0.83
History of Chronic Infections15 (13.51%)7 (5.30%)22 (9.05%)0.026
NSCLC Subtype
Adenocarcinoma93 (83.8%)101 (76.5%)194 (79.8%)0.366
Squamous Cell15 (13.5%)24 (18.2%)39 (16.0%)
Other3 (2.7%)7 (5.3%)10 (4.1%)
PD-L1 Activity 0.78
Absent39 (35.14%)48 (36.36%)87 (35.80%)
1–10%13 (11.71%)22 (16.67%)35 (14.40%)
11–20%6 (5.41%)5 (3.79%)11 (4.53%)
>20%37 (33.33%)38 (28.79%)75 (30.86%)
Unknown16 (14.41%)19 (14.39%)35 (14.40%)
Therapy 0.27
Monotherapy41 (36.94%)39 (29.55%)80 (32.92%)
Combination with Chemotherapy70 (63.06%)93 (70.45%)163 (67.08%)
Radiation Therapy27 (24.3%)38 (28.8%)65 (26.7%)0.469
Surgery 0.097
No Surgery90 (81.08%)95 (71.97%)185 (76.13%)
Primary Surgery9 (8.11%)23 (17.42%)32 (13.17%)
Secondary Surgery12 (10.81%)14 (10.61%)26 (10.70%)
Number of Cycles
Total (median)5.00 (2.00-13.00)8.00(4.00–12.00)7.00 (3.00–12.00)0.057
Distribution of cycles 0.067
120 (18.02%)11 (8.33%)31 (12.76%)
210 (9.01%)11 (8.33%)21 (8.64%)
≥381 (72.97%)110 (83.33%)191 (78.60%)
Line of Therapy 0.60
1L85 (76.58%)93 (70.45%)178 (73.25%)
2L21 (18.92%)31 (23.48%)52 (21.40%)
>2L5 (4.50%)8 (6.06%)13 (5.35%)
Antibiotics at time of ICI initiation21 (18.92%)8 (6.06%)29 (11.93%)0.002
GCSF Use12 (10.81%)16 (12.12%)28 (11.52%)0.75
ICI: Immune checkpoint inhibitor. T2DM: Type 2 diabestes mellitus; CAD: Coronary artery disease; COPD: Chronic obstructive pulmonary disease; ECOG: Eastern Cooperative Oncology Group; GCSF: Granulocyte colony stimulating factor. 1L: First line; 2L: second line.
Table 2. Distribution, frequency, timing of infections.
Table 2. Distribution, frequency, timing of infections.
1st Infection2nd Infection3rd Infection
Total1115518
Bacteremia8 (7.2%)3 (5.4%)0 (0.0%)
COVID-193 (2.7%)0 (0.0%)0 (0.0%)
Head and Neck5 (4.5%)3 (5.4%)0 (0.0%)
Intra-abdominal6 (5.4%)3 (5.4%)1 (5.6%)
Intracranial0 (0.0%)1 (1.8%)0 (0.0%)
Neutropenic Fever3 (2.7%)0 (0.0%)2 (11.1%)
Presumed Sepsis3 (2.7%)3 (5.4%)1 (5.6%)
Pulmonary48 (43.2%)28 (50.9%)9 (50.0%)
SSTI15 (13.5%)5 (9.1%)4 (22.2%)
Urinary 20 (18.0%)9 (16.4%)1 (5.6%)
Type of Pathogen
Bacterial90 (81.1%)50 (90.9%)15 (83.3%)
Virus18 (16.2%)2 (3.6%)2 (11.1%)
Fungal3 (2.7%)3 (5.4%)1 (5.6%)
Culture Data Available51 (45.9%)21 (38.2%)6 (33.3%)
Result of Infection
ED Visits6 (5.4%)3 (5.4%)0 (0.0%)
Inpatient Admission78 (70.3%)36 (65.4%)11 (61.1%)
IMU/ICU Admission13 (11.7%)14 (25.4%)6 (33.3%)
Outpatient14 (12.6%)3 (5.4%)1 (5.6%)
Median time to infection (days)58 (0–1465)124 (6–844)213.5 (25–1414)
SSTI: Skin and soft tissue infection.
Table 3. Univariable and Multivariable Analysis.
Table 3. Univariable and Multivariable Analysis.
UnivariableMultivariable
OR (95% CI)p-ValueOR (95% CI)p-Value
Age1.01 (0.99, 1.04)0.319
Female1.54 (0.93, 2.56)0.0961.52 (0.86, 2.70)0.153
T2DM0.88 (0.49, 1.59)0.674
CAD1.65 (0.91, 2.96)0.0961.53 (0.79, 2.95)0.204
COPD1.96 (1.15, 3.33)0.0131.59 (0.88, 2.85)0.121
Alcoholism0.58 (0.19, 1.74)0.327
Chronic indwelling catheter1y3.64 (0.37, 35.49)0.266
Smoking Status
NeverReference
Previous1.56 (0.73,3.32)0.251
Current1.02 (0.41,2.55)0.971
ECOG
0Reference Reference
13.23 (1.14, 9.10)0.0272.49 (0.86, 7.25)0.094
28.14 (2.50, 26.55)0.0014.85 (1.41, 16.70)0.012
34.6 (0.71, 29.84)0.111.80 (0.22, 14.79)0.583
4Empty Empty
Unknown6.07 (2.03, 18.20)0.0014.52 (1.44, 14.13)0.010
Previous ICI therapy0.89 (0.3, 2.63)0.827
Surgery
No SurgeryReference Reference
Primary Surgery0.41 (0.18, 0.94)0.0350.42 (0.17, 1.04)0.060
Secondary Surgery0.90 (0.40, 2.06)0.8121.02 (0.43, 2.43)0.966
Immunosuppressive therapy for irAE1.87 (1.0–3.51)0.057
History of Chronic Infections2.79 (1.09, 7.11)0.0321.74 (0.61, 4.97)0.304
Antibiotics/antivirals3.62 (1.53, 8.53)0.0033.17 (1.16, 8.66)0.024
GCSF use0.88 (0.4, 1.95)0.75
Line of therapy
1LReference
2L0.74 (0.40, 1.39)0.349
>2L0.68 (0.22, 2.17)0.519
Therapy
MonotherapyReference
Combination0.72 (0.42, 1.22)0.223
Number of cycles
1Reference Reference
20.5 (0.16–1.55)0.230.44 (0.12, 1.54)
3 or more0.41 (0.18–0.89)0.0250.42 (0.17, 1.02)
Table 4. Outcomes during pembrolizumab therapy. ED: Emergency Department.
Table 4. Outcomes during pembrolizumab therapy. ED: Emergency Department.
Infection
YesNoTotalp-Value
N = 111N = 132N = 243
Immune Related Adverse Events33 (29.73%)33 (25.00%)66 (27.16%)0.41
Colitis4 (3.60%)7 (5.30%)11 (4.53%)0.53
Adrenalitis0 (0.00%)1 (0.76%)1 (0.41%)0.36
Thyroiditis8 (7.21%)9 (6.82%)17 (7.00%)0.91
Pneumonitis17 (15.32%)7 (5.30%)24 (9.88%)0.009
Dermatitis9 (8.11%)13 (9.85%)22 (9.05%)0.64
Hepatitis3 (2.70%)0 (0.00%)3 (1.23%)0.057
Other8 (7.21%)5 (3.79%)13 (5.35%)0.24
ED Visit37 (33.33%)26 (19.70%)63 (25.93%)0.016
Inpatient87 (78.38%)53 (40.15%)140 (57.61%)<0.001
IMU/ICU admissions26 (23.42%)5 (3.79%)31 (12.76%)<0.001
Death74 (66.67%)70 (53.03%)144 (59.26%)0.031
Cause of Death 0.041
Primary Disease49 (66.22%)61 (87.14%)110 (76.39%)
Infection11 (14.86%)4 (5.71%)15 (10.42%)
Multiorgan Failure9 (12.16%)3 (4.29%)12 (8.33%)
Second Cancer1 (1.35%)0 (0.00%)1 (0.69%)
Other4 (5.41%)2 (2.86%)6 (4.17%)
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Burns, E.A.; Gee, K.; Kieser, R.B.; Xu, J.; Zhang, Y.; Crenshaw, A.; Muhsen, I.N.; Mylavarapu, C.; Esmail, A.; Shah, S.; et al. Impact of Infections in Patients Receiving Pembrolizumab-Based Therapies for Non-Small Cell Lung Cancer. Cancers 2023, 15, 81. https://doi.org/10.3390/cancers15010081

AMA Style

Burns EA, Gee K, Kieser RB, Xu J, Zhang Y, Crenshaw A, Muhsen IN, Mylavarapu C, Esmail A, Shah S, et al. Impact of Infections in Patients Receiving Pembrolizumab-Based Therapies for Non-Small Cell Lung Cancer. Cancers. 2023; 15(1):81. https://doi.org/10.3390/cancers15010081

Chicago/Turabian Style

Burns, Ethan A., Kelly Gee, Ryan B. Kieser, Jiaqiong Xu, Yuqi Zhang, Aubrey Crenshaw, Ibrahim N. Muhsen, Charisma Mylavarapu, Abdullah Esmail, Shivan Shah, and et al. 2023. "Impact of Infections in Patients Receiving Pembrolizumab-Based Therapies for Non-Small Cell Lung Cancer" Cancers 15, no. 1: 81. https://doi.org/10.3390/cancers15010081

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop