scholarly journals 16. Checking in with novel cancer therapies: a case of immunotherapy-related large vessel vasculitis

2019 ◽  
Vol 3 (Supplement_1) ◽  
Author(s):  
Yik Long Man ◽  
Neil Morton ◽  
Begoña Lopez

Abstract Introduction Immune checkpoint inhibition has revolutionised the management of patients with cancer. However, many immunotherapy-related adverse events have been recognised, such as colitis and dermatitis. We are increasingly aware of patients presenting to rheumatology with musculoskeletal complaints including polymyalgia-like symptoms or an inflammatory arthritis. More uncommonly, patients can present with large vessel vasculitis. We present a case of immunotherapy-related large vessel vasculitis following treatment with a combination of ipilimumab and nivolumab. Case description A 67-year-old man has been known to the oncology team with prostate cancer since 2014. He developed osteoblastic metastases despite androgen deprivation therapy and he was subsequently enrolled onto the NEPTUNE study which involved a combination of ipilimumab and nivolumab. Three weeks after his first cycle of immunotherapy, he developed fevers, diarrhoea and a macular rash. He was admitted for a flexible sigmoidoscopy and biopsies demonstrated inflammation in keeping with immunotherapy-related colitis. It was also noted that his thyroxine level was 64.7pmol/L with a TSH of 0.02mlU/L and this was thought to be immunotherapy-related thyroiditis. His immunotherapy was discontinued and he was on a weaning course of prednisolone with a good response.  Five months after his single cycle of immunotherapy, he began reporting generalised aches which were worse in his chest and radiated to his right scapula. He also had bilateral shoulder pain but no specific stiffness. This pain was more noticeable as he weaned off the prednisolone. He had no claudication, headaches or constitutional symptoms. A CT pulmonary angiogram showed no evidence of pulmonary emboli, but there was an incidental finding of circumferential thickening of the aorta suggestive of a vasculitis. Inflammatory markers were notably raised - ESR 127mm/h and CRP 199mg/L. There was no evidence of infection on cultures. He was referred to the rheumatology team. Examination was unremarkable with no evidence of weak pulses or bruits. Immunology tests were all negative. An urgent PET-CT was organised which demonstrated extensive active large vessel vasculitis involving the aorta, subclavian, axillary, carotid and vertebral arteries. He was given one dose of methylprednisolone (1mg/kg) which resulted in a marked improvement in his pain overnight. He received two further doses of methylprednisolone and his CRP improved to 38mg/L. He continues to improve on a weaning course of prednisolone.  Discussion Ipilimumab was the first checkpoint inhibitor approved for cancer in 2010. Immune checkpoint inhibitors have since become an expanding field in oncology, particularly in resistant or advanced cases of melanoma and lung cancer. There are currently six checkpoint inhibitors licensed by the US Food and Drug Administration. These are monoclonal antibodies targeting the checkpoint pathway including CTLA4, PD-1 and PDL-1. There are well documented case series with regard to immunotherapy-related toxicities including colitis, dermatitis and endocrinopathies. More relevant to rheumatologists, checkpoint inhibitors have also been associated with rheumatic presentations including inflammatory arthritis, polymyalgia rheumatica, sicca symptoms, myositis and vasculitis. A review of the literature in 2018 found 53 cases of vasculitis associated with checkpoint inhibition, of which 20 were confirmed. All these cases were resolved by withholding the immune checkpoint inhibitor and where necessary, giving steroid therapy. On the whole, immunotherapy-related vasculitis is not as common as arthritis or polymyalgia. As the use of checkpoint inhibitors becomes more widespread, it is important that as rheumatologists, we are aware of the various rheumatic conditions that they can trigger and how to manage them. Key learning points This single case highlights the wide range of immunotherapy-related adverse events associated with immune checkpoint inhibition. Their use in clinical practice will likely become more widespread owing to their success in treating a variety of advanced or resistant malignancies. Apart from being familiar with the various rheumatic complaints, we should also be aware of the other systems that can become involved, so that the patient is managed holistically. Symptoms will usually improve with termination of the checkpoint inhibitor but steroid therapy is often required. The addition of disease modifying anti-rheumatic drugs should be considered in cases where there are relapsing symptoms whilst weaning steroids. However, this is a decision that requires a multidisciplinary approach since it could affect the prognosis of the underlying malignancy. With more research into this area, there will a better understanding of the true incidence of immunotherapy-related adverse events in these patients and how to reduce these in the future. Conflict of interest The authors declare no conflicts of interest.

2020 ◽  
Vol 38 (6) ◽  
pp. 576-583 ◽  
Author(s):  
Hamzah Abu-Sbeih ◽  
David M. Faleck ◽  
Biagio Ricciuti ◽  
Robin B. Mendelsohn ◽  
Abdul R. Naqash ◽  
...  

PURPOSE The risk of immune checkpoint inhibitor therapy–related GI adverse events in patients with cancer and inflammatory bowel disease (IBD) has not been well described. We characterized GI adverse events in patients with underlying IBD who received immune checkpoint inhibitors. PATIENTS AND METHODS We performed a multicenter, retrospective study of patients with documented IBD who received immune checkpoint inhibitor therapy between January 2010 and February 2019. Backward selection and multivariate logistic regression were conducted to assess risk of GI adverse events. RESULTS Of the 102 included patients, 17 received therapy targeting cytotoxic T-lymphocyte antigen-4, and 85 received monotherapy targeting programmed cell death 1 or its ligand. Half of the patients had Crohn’s disease, and half had ulcerative colitis. The median time from last active IBD episode to immunotherapy initiation was 5 years (interquartile range, 3-12 years). Forty-three patients were not receiving treatment of IBD. GI adverse events occurred in 42 patients (41%) after a median of 62 days (interquartile range, 33-123 days), a rate higher than that among similar patients without underlying IBD who were treated at centers participating in the study (11%; P < .001). GI events among patients with IBD included grade 3 or 4 diarrhea in 21 patients (21%). Four patients experienced colonic perforation, 2 of whom required surgery. No GI adverse event–related deaths were recorded. Anti–cytotoxic T-lymphocyte antigen-4 therapy was associated with increased risk of GI adverse events on univariable but not multivariable analysis (odds ratio, 3.19; 95% CI, 1.8 to 9.48; P = .037; and odds ratio, 4.72; 95% CI, 0.95 to 23.53; P = .058, respectively). CONCLUSION Preexisting IBD increases the risk of severe GI adverse events in patients treated with immune checkpoint inhibitors.


2019 ◽  
Vol 25 (4) ◽  
pp. 954-960 ◽  
Author(s):  
Catherine E Renna ◽  
Elizabeth N Dow ◽  
Jason J Bergsbaken ◽  
Ticiana A Leal

Introduction The development of immune checkpoint inhibitors has revolutionized cancer treatment and is now a part of the treatment paradigm for several malignancies. Although immune checkpoint inhibitors are generally well tolerated, treatment is associated with immune-related adverse events, some serious and potentially life threatening. Early identification and prompt appropriate management of immune-related adverse events are crucial to prevent morbidity and mortality. The complexity and severity of immune-related adverse events require interdisciplinary collaboration to optimize care. Patient and caregiver education and continued communication between patients and members of the oncology care team are vital for timely recognition and successful management of immune-related adverse events. The objective of this program is to provide a proof of concept; a pharmacist-led immune checkpoint inhibitor management program will increase early recognition and management of immune-related adverse events through patient and caregiver education and proactively assessing patients for toxicities. Methods At the University of Wisconsin Carbone Cancer Center, we developed and implemented a pharmacist-driven program, referred to as the immune checkpoint inhibitor program, which aimed to ensure patient and caregiver education and continuous monitoring of immune-related adverse events. This program utilized pharmacist–patient encounters to improve patient and caregiver education and follow-up monitoring. The design and implementation are detailed. Pharmacist interventions and patient outcomes were evaluated. Results At interim analysis, 47 patients were enrolled in the program and pharmacists completed 34 interventions on 26 patients. Pharmacists are well positioned to educate patients and caregivers on immune checkpoint inhibitor therapy and provide proactive monitoring to detect immune-related adverse events. We hypothesize that the interventions made by pharmacist may lead to earlier recognition and treatment of immune-related adverse events.


Rheumatology ◽  
2018 ◽  
Vol 57 (4) ◽  
pp. 760-762 ◽  
Author(s):  
Jemima Albayda ◽  
Clifton O Bingham ◽  
Ami A Shah ◽  
Ronan J Kelly ◽  
Laura Cappelli

2019 ◽  
Vol 37 (8_suppl) ◽  
pp. 131-131 ◽  
Author(s):  
Lia Head ◽  
Nicholas Gorden ◽  
Robert Van Gulick ◽  
Carol M. Amato ◽  
Ashley Frazer-Abel ◽  
...  

131 Background: Immune-related adverse events (IRAE) occur commonly with immune checkpoint inhibitor therapy for the treatment of cancer, although the specific event and severity can vary widely. Little is known regarding factors that may predict which patients will develop an IRAE. The goal of this study is to identify blood biomarkers predictive of IRAE associated with immune checkpoint inhibitor therapy. Methods: Blood samples collected from patients with melanoma prior to receiving therapy with immune checkpoint inhibitors were obtained from the University of Colorado Skin Cancer Biorepository. Testing for a panel of autoantibodies and cytokines (ANA, CCP 3.1, IL-1 beta, IL-2, IL-6, IL-10, IL-12, IP-10, MCP-1, TNF alpha, IFN alpha 2, IFN gamma) in serum samples from patients who had at least one documented IRAE was performed by Exsera BioLabs. Descriptive statistics were used to evaluate biomarker levels in relation to type, grade, and number of adverse events. Results: Pre-treatment samples from 45 patients were evaluated. Median age was 55; 26 were male and 19 were female. The most common IRAEs were colitis (n = 22), thyroid dysfunction (n = 21), and dermatitis (n = 20). Most IRAEs were grade 2 in severity, and the majority of patients (n = 36) experienced more than 1 IRAE. TNF alpha was elevated in 60% of patient samples, while IFN alpha 2 was elevated in 44%. Borderline ANA was detected in 27% of samples and ANA was positive in 11%. No samples had elevation of IL-2. Between 9% and 18% of samples had elevation of the other immune markers tested (IFN gamma, IL-1 beta, IL-6, IL-10, IL-12, and CCP 3.1). Elevation of TNF alpha and IFN alpha 2 were associated with higher grades of IRAEs. No associations between immune markers and the number or type of adverse events in an individual patient were noted. Results from 15 patients who did not have a documented IRAE on immune checkpoint inhibitor therapy are currently pending to confirm these findings are unique to patients developing IRAE. Conclusions: This preliminary data suggests that baseline elevations of TNF alpha and IFN alpha 2 may predict development of IRAEs with immune checkpoint inhibitor therapy. Results from samples from patients who did not develop an IRAE on therapy will be reported at the meeting.


2020 ◽  
Vol 38 (4_suppl) ◽  
pp. 117-117
Author(s):  
M. Cecilia Monge B. ◽  
Changqing Xie ◽  
Seth M. Steinberg ◽  
Suzanne Fioraventi ◽  
Melissa Walker ◽  
...  

117 Background: The benefit of immune checkpoint inhibition is limited to the small percentage of advanced colorectal cancer (CRC) patients whose tumors present mismatch repair (MMR) gene abnormalities; immunotherapy has not shown benefit in patients with MMR proficient CRC. Oncolytic immunotherapy represents a unique therapeutic platform. This phase I trial tests the safety of the combination of pexastimogene devacirepvec (Pexa-Vec) plus durvalumab (durva) in patients with locally advanced or metastatic CRC. Methods: Eligible patients with advanced proficient mixed match repair (pMMR) CRC received intravenous infusion of Pexa-Vec at dose level 3 x 108 plaque forming units (pfu) (DL1) or at 109 pfu (DL2) every 2 weeks for 4 doses and durva 1500 mg every 28 days. Response was assessed with CT every 8 weeks. Adverse events were recorded and managed. The primary endpoint included safety, tolerability and feasibility of this combination therapy. Samples of tumor and peripheral blood were collected for assessment of immune parameters. Results: Sixteen patients (6 males and 10 females) enrolled with a median age of 52.1 years (range 39-69) from Dec, 2017 to Oct, 2018. Four patients were treated with Pexa-Vec at DL1 and durva;twelve patients were treated with Pexa-Vec at DL2 and durva.The most common treatment related adverse events (TRAE) included fever 15/16 (94 %), hypotension 12/16 (75 %), chills 12/16 (75%), fatigue 8/16 (50%), sinus tachycardia 7/16 (44%) and rash 6/16 (38%). Grade 3/4 TRAEs were reported in 8/16 (50%)patients; the most common were fever 7/16 (44 %), lymphopenia 2/16 (13%), neutropenia 1/16 (6%) and anemia 1/16 (6%). 14 patients were evaluable for response analysis; one patient 1/14 (7 %) achieved a confirmed partial response (lasting 7.1 months) and continues to receive treatment, while 13 patients had progressive disease. The median progression free survival (PFS) was 2.2 months (95% CI: 2.2-2.3 months) and the median overall survival (OS) was 7.5 months (CI: 4.9-10.1 months). Conclusions: Combination therapy of Pexa-Vec with durva ICI issafe, well tolerated and demonstrates possible activity in patients with advanced pMMR CRC. Clinical trial information: NCT03206073.


2019 ◽  
Vol 29 (5) ◽  
pp. 516-521 ◽  
Author(s):  
Victoria Grätz ◽  
Ewan A. Langan ◽  
Alexander Neumann ◽  
Detlef Zillikens ◽  
Patrick Terheyden

2019 ◽  
Vol 48 (3) ◽  
pp. 030006051988645 ◽  
Author(s):  
Yanmin Li ◽  
Zhengping Wang ◽  
Ting Guo ◽  
Shenghua Liu ◽  
Chenchen Feng

Background Immune checkpoint inhibition (ICI) represents a novel treatment modality for refractory cancers, and improving prediction of potential responders is critical. Method We hypothesized that ICI is a systemic-effecting mechanism. The objective response rate (ORR) for anti-PD-1, anti-PD-L1, anti-CTLA-4, or combination therapy was plotted against the corresponding all-grade and grade 3–4 (G3/4) treatment-related adverse events (TRAEs) across several cancer types using an extensive literature search (MEDLINE and Google Scholar; December 1, 2012–December 30, 2017). Results Sixty-six eligible studies comprised 76 cohorts and 25 cancer types. A significant correlation was present between all-grade or G3/4 TRAEs and the ORR. The correlation coefficient was 0.5 for all-grade and 0.4 for G3/4 TRAEs, suggesting that >50% of the differences in the ORR across cancer types may be reflected by TRAEs and 40% of ORR differences may be predicted by G3/4 TRAEs. Hodgkin’s lymphoma and Merkel cell carcinoma showed a better response, while adrenocortical cancer, breast cancer, and uveal melanoma showed a worse response, compared with that predicted by TRAE. Conclusion There is a strong relationship between TRAEs and ICI activity across multiple cancers. The toxicity profile compared with the ORR to ICIs should be investigated in phase I trials.


2020 ◽  
Vol 20 (7) ◽  
pp. 545-557
Author(s):  
Rohit Thummalapalli ◽  
Hanna A. Knaus ◽  
Ivana Gojo ◽  
Joshua F. Zeidner

Despite recent therapeutic advancements, acute myeloid leukemia (AML) remains a challenging clinical entity with overall poor outcomes. Given the evident role of T cell-mediated immunity in response to allogeneic stem cell transplantation and donor lymphocyte infusions, strategies that enhance immune activation and mitigate immune dysfunction represent attractive therapeutic platforms to improve clinical outcomes in AML. Pre-clinical data suggest that immune dysfunction is a major contributor to AML progression and relapse. Increased expression of immune checkpoints such as programmed death 1 (PD-1) contributes to AML immune evasion and is associated with disease progression. Immune checkpoint inhibition is being explored in AML with early evidence of clinical activity, particularly in combination with cytotoxic chemotherapy and hypomethylating agents. In this review, we explore the scientific rationale behind the use of immune checkpoint inhibition either as single agents or in combination with hypomethylating agents or cytotoxic chemotherapy and provide a clinical update of both completed and ongoing trials in AML.


Sign in / Sign up

Export Citation Format

Share Document