Summary
CDX-014 is an antibody-drug conjugate directed against TIM-1, a surface marker highly expressed in renal cell carcinoma (RCC) and ovarian carcinoma. This phase I, first-in-human trial was conducted to evaluate the safety and preliminary activity of CDX-014 in patients with advanced refractory RCC, following a dose-escalation and dose expansion design. CDX-014 was administered intravenously at doses ranging from 0.15 to 2.0 mg/kg every 2 or 3 weeks until progression or unacceptable toxicity. Sixteen patients received at least one dose of CDX-014. The maximum tolerated dose was not identified. Most frequent adverse grade 1 or 2 adverse events included nausea (38%), fatigue, alopecia, elevation of AST and decreased appetite (25% each). Adverse events of grade 3 or more included hyperglycemia (19%), urosepsis (6%), and one multi-organ failure (6%) responsible for one treatment-related death. Two patients discontinued therapy for adverse events including fatigue grade 2 and urosepsis grade 4. CDX-014 showed antitumor activity with one prolonged partial response and a clinical benefit rate (objective response or stable disease >6 months) of 31%. The two patients that exhibited the most marked tumor shrinkage had high TIM-1 expression on tumor tissue. Overall, CDX-014 exhibited a manageable toxicity profile and early signs of activity, supporting further evaluation of antibody-drug conjugates in patients with advanced RCC and potentially other TIM-1 expressing cancers. Trial registration https://clinicaltrials.gov/ct2/show/NCT02837991 NCT02837991; July 20, 2016.
Keywords: Renal cell carcinoma, Antibody-drug conjugate, Phase 1 trial, Salvage therapy, First-in-human, Metastatic renal cell carcinoma
Introduction
Outcomes of patients with renal cell carcinoma (RCC) have improved since the advent of antiangiogenic therapies and immune checkpoint inhibitors; with combinations of immunotherapies alone or with vascular-endothelial growth factor receptor (VEGFR) inhibitors approved in the first-line setting given phase 3 data showing superiority over the tyrosine kinase inhibitor (TKI) sunitinib [1-3]. Most patients with metastatic disease will eventually progress on therapy. Unfortunately, therapeutic options are limited after failure of VEGFR-targeted therapies and immune checkpoint inhibitors, and identification of new therapeutic targets is essential.
Antibody-drug conjugates allow for ability to target high concentrations of cytotoxic chemotherapy to tumor sites using a monoclonal antibody as a delivery vehicle. They are emerging as a new standard of care in other genitourinary malignancies with drugs such as enfortumab vedotin targeting nectin-4 universally expressed on urothelial carcinoma, recently approved by the FDA in December 2019 [4]. Similar target antigens in RCC are needed. T cell ImmunoglobulinMucin-1 (TIM-1), also known as Kidney Injury Molecule 1 (KIM-1), is a transmembrane glycoprotein that is upregulated in tubular cells following ischemia [5]. Expression of TIM-1 notably allows tubular cells to detect and phagocytize apoptotic bodies in the context of kidney injury [6]. Considering that such tubular cells are involved in specific dedifferentiation programs that favor proliferation in response to acute kidney injury, TIM-1 has also been considered as a marker of dedifferentiated tubular cells [7]. Recent insights demonstrated that TIM-1 was detectable in tumor tissue in more than 70% of patients with clear cell or papillary RCC, consistent with dedifferentiation and proliferation processes in tumor cells arising from the proximal tubule [8]. Conversely, TIM-1 expression levels were low in normal renal cortex (10%), as well as other solid solid tumors [9]. Therefore, TIM-1 has been considered as a selective biomarker for RCC diagnosis, as well as a potential putative target for an ADC therapeutic approach.
CDX-014 is an antibody-drug conjugate (ADC) consisting of a human immunoglobulin G1 directed against TIM-1, covalently linked with the antimitotic monomethyl auristatin E (MMAE) via standard maleimide conjugation [10]. CDX-014 binds to TIM-1 expressing cells and upon internalization, proteolysis of the valine-citrulline linker releases MMAE, leading to cytotoxic death through inhibition of tubulin polymerization [11]. In vitro and in vivo activity has been demonstrated in TIM-1 expressing tumor cell lines [11]. Herein we evaluate CDX-014 in a phase I trial in patients with advanced RCC who failed prior VEGFR-directed therapy.
Materials and methods
Patients
Patients with advanced clear cell or papillary RCC who experienced progression after at least two lines of systemic therapy, including at least one TKI were included. Additional main inclusion criteria included measurable disease by Response Criteria in Solid Tumors (RECIST) 1.1 criteria, a Karnofsky performance status ≥70%, and adequate renal, hepatic, and hematological laboratory values. Archival or pre-treatment formalin-fixed paraffin embedded (FFPE) tumor tissue were required for TIM-1 expression assessment. Patients with history of brain metastases were excluded unless previously treated and asymptomatic for at least 2 months. The full list of inclusion criteria is availiable at https://clinicaltrials.gov/ct2/show/NCT02837991.
Procedures
This trial was designed as a multicenter phase I, open-label, single arm study, and included a dose escalation phase followed by a dose expansion phase. Patients would be treated with one of the following doses of intravenous CDX-014: 0.15, 0.30, 0.60, 1.2, 1.8, 2.0, 2.2, and 2.4 mg/kg of body weight every 3 weeks. After 14 patients enrolled, protocol was amended to treat with escalating doses of 1.0, 1.2, 1.4, 1.6 and 1.8 mg/kg every 2 weeks. Treatment was administered until radiological progression, unacceptable toxicity, pregnancy, withdrawal of consent, decision of discontinuation by the physician, or death. Dose reductions were allowed following guidelines provided in the Data Supplement.
Toxicity was recorded at each visit according to the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE) v4.0. Disease assessments were performed every 6 weeks for 6 months, then every 8 weeks by computed tomography of the chest, abdomen, and pelvis, according to the RECIST 1.1 guidelines. Pharmacokinetic assessments were used to monitor exposure to free MMAE levels at each planned visit. TIM-1 expression levels were analyzed on archival or pre-treatment tumor tissue by immunohistochemistry and scored according to staining intensity and percentage of cells expressing TIM-1.
This trial was approved by the Institutional Review Boards of each participating institution and conducted in accordance with the principles of the Declaration of Helsinki and the International Conference on Harmonization.
Study objectives
The primary objective of the dose escalation phase was to determine the maximum tolerated dose (MTD) of CDX-014 in advanced refractory RCC. The primary objective of the dose expansion phase was to determine preliminary antitumor activity of CDX-014 at the MTD in this population as measured by the objective response rate (ORR).
Secondary objectives included tolerability, pharmacokinetics, and additional efficacy assessments: clinical benefit rate, duration of response, progression-free survival, and overall survival. Exploratory objectives included determination of TIM-1 expression in tissue and correlations with outcomes.
Clinical benefit rate was defined as the rate of complete response (CR), partial response (PR) and stable disease (SD) of at least 6 months. PFS was defined as time from therapy initiation to progression or death. Patients who are event-free were censored at the date of last evaluation for PFS. Overall survival (OS) was defined as time from therapy initiation to death or last contact for patients alive.
Statistical design
In the dose escalation phase, a minimum of 6 patients would be treated at the MTD prior to dose expansion. The first four dose levels of CDX-014 (0.15 to 1.2 mg/kg every 3 weeks) were to include a single patient each, and dose escalation would proceed in the absence of dose-limiting toxicity (Data Supplement). Further dose levels as would follow a standard 3 + 3 dose escalation scheme. A total of 56 patients were expected to be enrolled in the dose escalation phase. The dose expansion phase would include approximately 15 patients evaluable for response, allowing an estimation of the ORR with a 95% confidence interval (95%CI) width not greater than 0.52 based on the Clopper Pearson method.
All patients who receive at least one dose of CDX-014 were evaluable for safety. Patients also had at least one post-baseline disease assessment, or discontinued therapy due to death or clinical deterioration.
Descriptive statistics were used to report baseline characteristics of the population, response data, and safety analyses. Median and range were used to describe quantitative data, frequency, and proportion for qualitative data. Survival analyses were performed using Kaplan-Meier estimators and reported with 95% CIs. For exploratory biomarker analyses, patients were divided into two groups based on median TIM-1 expression on tumor tissue; Fisher and log-rank tests were respectively used to compare response rates and survival according to TIM-1 expression. All analyses were performed using NCSS Statistical Software (2019), NCSS, LLC. Kaysville, Utah.
Results
Patients
Sixteen patients were enrolled in the study and received at least one dose of CDX-014 between 7/20/2016 to 3/21/2018. Baseline characteristics of the patients are reported in Table 1. Patients were predominantly male (94%), with a median age of 67 years, and most (57%) had a Karnofsky performance status of 90% or 100%. Most patients had clear cell RCC (14/16, 88%), while 2/16 (13%) had papillary RCC, and 75% (12/16) belonged to intermediate or poor risk prognostic groups according to the IMDC criteria. The study population was heavily pretreated, with a median of 4 previous systemic therapies: VEGFR-directed therapies in all patients, immune checkpoint inhibitors in 94%, and mTOR inhibitors in 44%. Median followup was 15.2 months (95%CI 12.9–17.6). Twelve (75%) patients experienced progression or death at data cutoff on 11/26/2018, three did not experience progression and were still alive on therapy, and one was lost to follow-up.
Table 1.
Baseline characteristics of the patients
| Characteristic | Patients (N = 16) |
|---|---|
| Median age, years (range) | 67 (56–84) |
| Sex | |
| Female | 1 (6%) |
| Male | 15 (94%) |
| Histology | |
| Clear-cell | 14 (88%) |
| Papillary | 2 (13%) |
| Median number of previous therapies (range) | 4 (2–9) |
| Type of previous therapies | |
| Immune checkpoint inhibitor | 15 (94%) |
| VEGF/VEGFR-directed therapy | 16 (100%) |
| MTOR inhibitor | 7 (44%) |
| Karnofsky performance status | |
| 70 | 2 (13%) |
| 80 | 5 (31%) |
| 90 | 7 (44%) |
| 100 | 2 (13%) |
| IMDC risk groups | |
| Favorable | 4 (25%) |
| Intermediate | 8 (50%) |
| Poor | 4 (25%) |
Risk groups are defined according to the following risk factors: time from diagnosis to initiation of systemic therapy <1 year, hemoglobin < lower limit of normal, neutrophils > upper limit of normal, platelets > upper limit of normal, corrected calcium > upper limit of normal, Karnofsky performance status <80%
IMDC International metastatic renal cell carcinoma database
Safety
Patients were enrolled at increasing dose levels up to 2.0 mg/kg every 3 weeks and 1.2 mg/kg every 2 weeks (Fig. 1). Patients received a median of 4 cycles (range 1–28) of CDX-014 (Supplementary Table 1). Discontinuation of CDX-014 development led to early termination of the trial. The MTD of CDX-014 has not been reached at assessed dose levels.
Fig. 1.
CONSORT diagram and Enrollment summary
There were two dose-limiting toxicities, both occurring at a dose level of 2.0 mg/kg every 3 weeks. One patient experienced grade 3 rash, grade 3 liver dysfunction, grade 4 neutropenia, and ultimately grade 5 fatal multi-organ failure, after receiving one dose of CDX-014. Of note, this grade 5 adverse event occurred in a context of unrelated grade 1 acute kidney injury prior to the event, probably due to dehydration and/or administration of iodine-based radiocontrast. A second patient experienced a dose limiting toxicity of grade 3 hyperglycemia in the context of known diabetes mellitus at this dose level, which resolved with appropriate supportive therapy. The study protocol was amended to allow further dose exploration despite this second dose-limiting toxicity.
Most common adverse events were nausea (6/16, 38%), fatigue, alopecia, elevation of AST, and decreased appetite (4/16, 25% each), all of which were grade 1 or 2. In addition to previously mentioned adverse events, other grade 3 or 4 toxicities included grade 3 hyperglycemia (3/16, 19%) in patients treated at 1.2, 1.8 and 2.0 mg/kg, as well as one occurrence of grade 4 urosepsis after 5 doses of CDX-014 at 1.8 mg/kg in a patient treated with history of multiple urinary tract infections (Table 2). Two patients discontinued therapy with CDX-014 for toxicity: one for grade 4 urosepsis as described above, and one for grade 2 fatigue.
Table 2.
Treatment-related adverse events
| Adverse event | Grade 1–2 | Grade 3–4 | Total |
|---|---|---|---|
| Nausea | 6 (38%) | 0 | 6 (38%) |
| Fatigue | 4 (25%) | 0 | 4 (25%) |
| Alopecia | 4 (25%) | NA | 4 (25%) |
| AST Increased | 4 (25%) | 0 | 4 (25%) |
| Decreased Appetite | 4 (25%) | 0 | 4 (25%) |
| Rash | 2 (13%) | 1 (6%) | 3 (19%) |
| Neuropathy | 3 (19%) | 0 | 3 (19%) |
| ALT Increased | 3 (19%) | 0 | 3 (19%) |
| Diarrhea | 3 (19%) | 0 | 3 (19%) |
| Hyperglycemia | 0 | 3 (19%) | 3 (19%) |
| Neutropenia | 1 (6%) | 1 (6%) | 2 (13%) |
| Blood Creatinine Increased | 2 (13%) | 0 | 2 (13%) |
Reported adverse events occurred in at least 10% of patients. Data presented as N (%). One patient died from multi-organ failure, potentially treatment-related, after one dose of CDX-014 at 2.0 mg/kg
Pharmacokinetics
Free MMAE levels in peripheral blood were assessed at cycle 1 (Supplementary Table 2) and following cycles. There was a trend towards correlation between CDX-014 dosing and free MMAE Cmax after cycle 1 (Spearmann r = 0.48, p = 0.08). Free MMAE Cmax after cycle 1 was highest in the study patient who experienced grade 5 multi-organ failure suggestive of poor renal clearance of the MMAE [12].
Activity
CDX-014 activity was ultimately assessed in all patients who received at least one dose of therapy. One patient (6%) treated at 0.3 mg/kg every 3 weeks exhibited PR as best overall response (Fig. 2), ongoing after 17 months on therapy; this patient had been on single agent PD-1 inhibition prior to initiating therapy with CDX-014. Five patients (31%) exhibited clinical benefit from CDX-014. PFS and OS were 2.7 months (95%CI 1.2–8.0) and 12.6 months (95%CI 5.7–12.6), respectively.
Fig. 2.
Best overall RECIST response to CDX-014. CDX-014 dose [mg/kg] and TIM-1 expression [% tumor cells positive] are displayed in parenthesis for each patient. All patients are Q3W schedule unless noted as Q2W
Expression of TIM-1 and response to therapy
Most patients (14/16, 88%) had detectable expression of TIM-1 on tumor cells from archival tissue. There was no statistical correlation between expression of TIM-1 and response, clinical benefit, or survival. However, both patients with the largest decrease in tumor burden had strong TIM-1 expression, including one patient with a prolonged PR and one patient with SD that experienced 29% tumor shrinkage (Fig. 3).
Fig. 3.
Response by “spider plots”
Discussion
CDX-014 demonstrated a manageable safety profile and modest evidence of antitumor activity in this highly pretreated RCC population, with 3/16 pts. (19%) remaining alive without progression at data cutoff. The observed toxicity profile is compatible with those of ADCs, including chemotherapy-like adverse events including fatigue, nausea, and cytopenia. Hyperglycemia was recorded in 3 patients (19%), and may represent a class-dependent adverse event though at this time, the mechanism is unclear.
There was one event of multi-organ failure of unknown origin deemed related to the study drug. TIM-1 is notably overexpressed not only in RCC tissue, but also in regenerating tubular cells in a context of acute kidney injury [13], and it has been suggested that measuring the level of TIM-1 in the urine can be a sensitive test for the early diagnosis of acute kidney injury [14]. TIM-1 expressing tubular epithelial cells phagocytose apoptotic cells to reduce inflammation and innate immune response, but its expression is also involved in the repair process where it promotes migration of renal tubular epithelial cells [15, 16]. The fatal adverse event occurred after one dose of treatment in a patient who experienced initial grade 1 acute kidney injury in the setting of recent CT scan with contrast. While it is unknown whether this patient had increased expression of TIM-1 in their normal kidney, increased TIM-1 may have led to increased binding of CDX-014 to non-neoplastic kidney tissue, begetting further kidney injury and ultimately multisystem organ failure and death similar to what is seen with a hyperacute rejection to an allogeneic renal transplant [17]. Unfortunately, autopsy was not able to be performed. The high level of free MMAE in this patient was also a likely contributing factor to the toxicity.
Grade 3 or 4 hyperglycemia was noted in 3 patients, and the mechanism for this remains unclear but it is seen with other antibody drug conjugates such as enfortumab vedotin which utilize the same cytotoxic payload MMAE [4, 18]. These toxicities highlight the need to find RCC-specific biomarkers, especially as ADCs exhibit prolonged half-life compared to small molecules, but shorter than unconjugated antibodies [19].
Further biomarker studies are needed to identify appropriate ADC targets in the context of RCC. TIM-1 plays a role outside of the kidney, notably in immune dysfunction diseases such as systemic lupus erythematosus, and it has been linked with Th2 immune response in asthma [18, 20]. As such, with the pivotal role immunotherapy now plays in the management of RCC, there is potential for significant interactions with preceding therapies.
Major advances have been made with ADCs in other genitourinary malignancies, primarily enfortumab-vedotin [4, 21] and sacituzumab-govitecan [22] in urothelial carcinoma, which demonstrated response rates of 55% and 31%, respectively, in highly refractory patients. Other ADCs have been investigated in RCC. The CD70-targeted ADC SGN-75 administered every 3 weeks showed prolonged disease control in up to 44% of patients with CD70-positive refractory RCC (N = 32) [23] though no further studies have been performed to date. The ENPP3-targeted ADC AGS-16C3F showed responses in 23% of patients with refractory RCC at 1.8 mg/kg (N = 13) [24]. This led to a phase 2 trial (NCT02639185) comparing AGS-16C3F to axitinib in patients with ccRCC or nccRCC expressing ENPP-3 which has completed accrual. Notably, unlike CDX-014, both SGN-75 and AGS-16C3F employ a non-cleavable linker between antibody and drug payload.
The success of ADCs in urothelial carcinoma is intriguing, but this is in the setting of a disease where cytotoxic chemotherapy remains the standard; combination of cisplatin with gemcitabine can achieve ORR approaching 50% [25]. While there is clearly a role for cytotoxic chemotherapy in the management of certain RCC histologies such as medullary carcinoma or collecting duct, there is minimal role in metastatic ccRCC. Chemotherapy plays a role in in aggressive subsets of RCC, including notably RCC with sarcomatoid dedifferentiation [26], where small studies have shown efficacy for doxorubicin and gemcitabine [27] or the combination of gemcitabine with sunitinib [28] in those patients, though of limited duration. Our data using a targeted approach, though limited, suggest delivering high doses of chemotherapy with an ADC targeting TIM-1 showed signs of anti-tumor activity.
Bringing new therapeutic avenues to RCC is essential, as options after failure of immune checkpoint inhibitors and VEGFR-directed TKIs are limited. CDX-014 is an ADC targeting TIM-1, a novel marker in RCC. The data was ultimately insufficient to make any judgement based on clinical activity. There are ongoing discussions with multiple companies to continue development of CDX- 014. In examining the relationship of high TIM-1 expression to response and response duration, there did appear to be a relationship between these parameters and high TIM-1 expression, however, a group of patients with similar high expression in their tumors experienced rapid progression on treatment (Figs. 2 and 3). This suggests that simply having high TIM-1 expression is insufficient for efficacy, with other factors such as cellular sensitivity to and intratumoral disposition of MMAE playing a potential role. Other ADCs are being explored, though it remains unknown how they will interact with approved therapy, including TKIs and immunotherapies, or if the chemotherapy delivered will have a beneficial effect given inherent resistance of ccRCC to cytotoxic chemotherapy. Furthermore, the dynamic nature and heterogeneity of target expression may affect ADC efficacy and would have to be specifically explored in RCC. Identification of specific targets could may also be relevant to the development of bispecific antibodies that could be used as immunotherapy strategies to direct immune cells towards tumor cells, aiming at overcoming mechanisms of resistance to conventional immune checkpoint inhibitors [29].
Conclusion
This first-in-human trial of CDX-014 demonstrated a novel approach to the management of refractory RCC. CDX-014 had a manageable safety profile and some patients experienced anti-tumor activity. Ongoing work is necessary to determine its clinical activity and optimal dosing strategy.
Supplementary Material
Acknowledgements
We thank the patients and their families for participating in this trial.
We thank Kevin Pels, PhD of Dana-Farber Cancer Institute for manuscript editing.
Funding information CellDex Therapeutics, Inc.
Supported by the following grants: CA 014089 (USC Norris CCSG).
Footnotes
Electronic supplementary material The online version of this article (https://doi.org/10.1007/s10637-020-00945-y) contains supplementary material, which is available to authorized users.
Conflict of interest BAM: discloses payment for consulting with Bayer, Astellas, Astra Zeneca, Seattle Genetics, Exelixis, Nektar, Pfizer, Janssen, Genentech and EMD Serono. He received research support to Dana Farber Cancer Institute (DFCI) from Bristol Myers Squibb, Calithera, Exelixis, Seattle Genetics.
RF: no competing interests.
MG reports other from Medimmune, other from Merck, other from BMS, other from Amgen, other from Tesaro, other from Beigene, other from ABBVIE, other from Aeglea, personal fees and other from Agenus, other from Arcus, other from Astex, other from BluePrint, other from Calithera, other from CellDex, other from Corcept, other from Clovis, other from Eli Lilly, other from Endocyte, other from Five Prime, other from Genocea, other from Neon, other from Plexxicon, personal fees and other from Imaging Endpoints, other from Revolution Medicine, other from Seattle Genetics, other from Serono, other from SynDevRx, other from Toleron, personal fees and other from Tracon, personal fees and other from Deciphera, personal fees and other from Salarius, outside the submitted work.
NA discloses payment for consultancy to Astellas, Astra Zeneca, BMS, Bayer, Clovis, Eisai, Exelixis, EMD Serono, Ely Lilly, Foundation Medicine, Genentech, Janssen, Merck, Novartis, Nektar, Pfizer, Pharmacyclics.
SG reports grants and personal fees from Bayer, grants and personal fees from BMS, grants from Novartis, personal fees from Exelixis, personal fees from Janssen, grants and personal fees from Corvus, personal fees from Genentech, personal fees from Sanofi/ Genzyme, grants and personal fees from Pfizer, grants from Acceleron, grants from Merck, grants from Agensys, grants from Eisai, personal fees from EMD Serono, outside the submitted work.
DIQ discloses payment for consulting with Bayer, BMS, Exelixis, Merck, Novartis and Pfizer.
MR, TH and TK own stock and are employees of Celldex Therapeutics, Inc.
TKC discloses payment for consulting to Analysis Group, AstraZeneca, Alexion, Sanofi/Aventis, Bayer, Bristol-Myers Squibb/ER Squibb and sons LLC, Cerulean, Eisai, Foundation Medicine Inc., Exelixis, Genentech, Roche, Roche Products Limited, F. Hoffmann-La Roche, GlaxoSmithKline, Heron Therapeutics, Lilly, Merck, Novartis, Peloton, Pfizer, EMD Serono, Prometheus Labs, Corvus, Ipsen, Up-to- Date, NCCN, Analysis Group, Michael J. Hennessy (MJH) Associates, Inc. (Healthcare Communications Company with several brands such as OnClive, PeerView and PER), L-path, Kidney Cancer Journal, Clinical Care Options, Platform Q, Navinata Healthcare, Harborside Press, American Society of Medical Oncology, NEJM, Lancet Oncology. He received research support to DFCI from Analysis Group, AstraZeneca, Alexion, Bayer, Bristol Myers-Squibb/ER Squibb and sons LLC, Cerulean, Eisai, Foundation Medicine Inc., Exelixis, Ipsen, Tracon, Genentech, Roche, Roche Products Limited, F. Hoffmann-La Roche, GlaxoSmithKline, Lilly, Merck, Novartis, Peloton, Pfizer, Prometheus Labs, Corvus, Calithera, Sanofi/Aventis, Takeda.
Ethical approval All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards.
Informed consent Informed consent was obtained from all individual participants included in the study.
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
References
- 1.Motzer RJ, Tannir NM, McDermott DF, Arén Frontera O, Melichar B, Choueiri TK, Plimack ER, Barthélémy P, Porta C, George S, Powles T, Donskov F, Neiman V, Kollmannsberger CK, Salman P, Gurney H, Hawkins R, Ravaud A, Grimm M-O, Bracarda S, Barrios CH, Tomita Y, Castellano D, Rini BI, Chen AC, Mekan S, McHenry MB, Wind-Rotolo M, Doan J, Sharma P, Hammers HJ, Escudier B (2018) Nivolumab plus Ipilimumab versus Sunitinib in advanced renal-cell carcinoma. N Engl J Med 378: 1277–1290. 10.1056/NEJMoa1712126 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Rini BI, Plimack ER, Stus V, Gafanov R, Hawkins R, Nosov D, Pouliot F, Alekseev B, Soulières D, Melichar B, Vynnychenko I, Kryzhanivska A, Bondarenko I, Azevedo SJ, Borchiellini D, Szczylik C, Markus M, McDermott RS, Bedke J, Tartas S, Chang Y-H, Tamada S, Shou Q, Perini RF, Chen M, Atkins MB, Powles T (2019) Pembrolizumab plus Axitinib versus Sunitinib for advanced renal-cell carcinoma. N Engl J Med 380:1116–1127. 10.1056/NEJMoa1816714 [DOI] [PubMed] [Google Scholar]
- 3.Motzer RJ, Penkov K, Haanen J, Rini B, Albiges L, Campbell MT, Venugopal B, Kollmannsberger C, Negrier S, Uemura M, Lee JL, Vasiliev A, Miller WH, Gurney H, Schmidinger M, Larkin J, Atkins MB, Bedke J, Alekseev B, Wang J, Mariani M, Robbins PB, Chudnovsky A, Fowst C, Hariharan S, Huang B, di Pietro A, Choueiri TK (2019) Avelumab plus Axitinib versus Sunitinib for advanced renal-cell carcinoma. N Engl J Med 380:1103–1115. 10.1056/NEJMoa1816047 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Rosenberg JE, O’Donnell PH, Balar AV, McGregor BA, Heath EI, Yu EY, Galsky MD, Hahn NM, Gartner EM, Pinelli JM, Liang S-Y, Melhem-Bertrandt A, Petrylak DP (2019) Pivotal trial of enfortumab vedotin in urothelial carcinoma after platinum and anti-programmed death 1/programmed death ligand 1 therapy. J Clin Oncol 37:2592–2600. 10.1200/JCO.19.01140 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Han WK, Bailly V, Abichandani R, Thadhani R, Bonventre JV (2002) Kidney injury Molecule-1 (KIM-1): a novel biomarker for human renal proximal tubule injury. Kidney Int 62:237–244. 10.1046/j.1523-1755.2002.00433.x [DOI] [PubMed] [Google Scholar]
- 6.Bonventre JV (2009) Kidney injury molecule-1 (KIM-1): a urinary biomarker and much more. Nephrol Dial Transplant 24:3265–3268. 10.1093/ndt/gfp010 [DOI] [PubMed] [Google Scholar]
- 7.Berger K, Moeller MJ (2014) Mechanisms of epithelial repair and regeneration after acute kidney injury. Semin Nephrol 34:394–403. 10.1016/j.semnephrol.2014.06.006 [DOI] [PubMed] [Google Scholar]
- 8.Han WK, Alinani A, Wu C-L, Michaelson D, Loda M, McGovern FJ, Thadhani R, Bonventre JV (2005) Human kidney injury molecule-1 is a tissue and urinary tumor marker of renal cell carcinoma. J Am Soc Nephrol 16:1126–1134. 10.1681/ASN.2004070530 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Lin F, Zhang PL, Yang XJ, Shi J, Blasick T, Han WK, Wang HL, Shen SS, Teh BT, Bonventre JV (2007) Human kidney injury molecule-1 (hKIM-1): a useful immunohistochemical marker for diagnosing renal cell carcinoma and ovarian clear cell carcinoma. Am J Surg Pathol 31:371–381. 10.1097/01.pas.0000213353.95508.67 [DOI] [PubMed] [Google Scholar]
- 10.Polakis P (2016) Antibody drug conjugates for cancer therapy. Pharmacol Rev 68:3–19. 10.1124/pr.114.009373 [DOI] [PubMed] [Google Scholar]
- 11.Thomas LJ, Vitale L, O’Neill T, Dolnick RY, Wallace PK, Minderman H, Gergel LE, Forsberg EM, Boyer JM, Storey JR, Pilsmaker CD, Hammond RA, Widger J, Sundarapandiyan K, Crocker A, Marsh HC, Keler T (2016) Development of a novel antibody–drug conjugate for the potential treatment of ovarian, lung, and renal cell carcinoma expressing TIM-1. Mol Cancer Ther 15:2946–2954. 10.1158/1535-7163.MCT-16-0393 [DOI] [PubMed] [Google Scholar]
- 12.Zhao B, Chen R, O’Connor OA, Gopal AK, Ramchandren R, Goy A, Matous JV, Fasanmade AA, Manley TJ, Han TH (2016) Brentuximab vedotin, an antibody–drug conjugate, in patients with CD30-positive haematologic malignancies and hepatic or renal impairment. Br J Clin Pharmacol 82:696–705. 10.1111/bcp.12988 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Zhang Z, Humphreys BD, Bonventre JV (2007) Shedding of the urinary biomarker kidney injury Molecule-1 (KIM-1) is regulated by MAP kinases and Juxtamembrane region. J Am Soc Nephrol 18: 2704–2714. 10.1681/ASN.2007030325 [DOI] [PubMed] [Google Scholar]
- 14.Jin Y, Shao X, Sun B, Miao C, Li Z, Shi Y (2017) Urinary kidney injury molecule-1 as an early diagnostic biomarker of obstructive acute kidney injury and development of a rapid detection method. Mol Med Rep 15:1229–1235. 10.3892/mmr.2017.6103 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Zhao X, Jiang C, Olufade R, Liu D, Emmett N (2016) Kidney injury Molecule-1 enhances endocytosis of Albumin in renal proximal tubular cells: KIM-1 expression and tubular albumin handling. J Cell Physiol 231:896–907. 10.1002/jcp.25181 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Zhang Z, Cai CX (2016) Kidney injury molecule-1 (KIM-1) mediates renal epithelial cell repair via ERK MAPK signaling pathway. Mol Cell Biochem 416:109–116. 10.1007/s11010-016-2700-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Nankivell BJ, Alexander SI (2010) Rejection of the kidney allograft. N Engl J Med 363:1451–1462. 10.1056/NEJMra0902927 [DOI] [PubMed] [Google Scholar]
- 18.Ueno T, Habicht A, Clarkson MR, Albin MJ, Yamaura K, Boenisch O, Popoola J, Wang Y, Yagita H, Akiba H, Ansari MJ, Yang J, Turka LA, Rothstein DM, Padera RF, Najafian N, Sayegh MH (2008) The emerging role of T cell Ig mucin 1 in alloimmune responses in an experimental mouse transplant model. J Clin Invest 118:742–751. 10.1172/JCI32451 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Hedrich WD, Fandy TE, Ashour HM, Wang H, Hassan HE (2018) Antibody–drug conjugates: pharmacokinetic/Pharmacodynamic modeling, preclinical characterization, clinical studies, and lessons learned. Clin Pharmacokinet 57:687–703. 10.1007/s40262-017-0619-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.de Souza AJ, Oak JS, Jordanhazy R, DeKruyff RH, Fruman DA, Kane LP (2008) T cell Ig and Mucin Domain-1-mediated T cell activation requires recruitment and activation of Phosphoinositide 3-kinase. J Immunol 180:6518–6526. 10.4049/jimmunol.180.10.6518 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Petrylak DP, Balar AV, O’Donnell PH, McGregor BA, Heath EI, Yu EY, Galsky MD, Hahn NM, Gartner EM, Pinelli J, Melhem-Bertrandt A, Rosenberg JE (2019) EV-201: results of enfortumab vedotin monotherapy for locally advanced or metastatic urothelial cancer previously treated with platinum and immune checkpoint inhibitors. J Clin Oncol 37:4505–4505. 10.1200/JCO.2019.37.18_suppl.LBA4505 [DOI] [Google Scholar]
- 22.Tagawa ST, Faltas BM, Lam ET, Saylor PJ, Bardia A, Hajdenberg J, Morgans AK, Lim EA, Kalinsky K, Simpson PS, Galsky MD, Goswam T, Wegener WA, Petrylak DP (2019) Sacituzumab govitecan (IMMU-132) in patients with previously treated metastatic urothelial cancer (mUC): results from a phase I/II study. J Clin Oncol 37:354–354. 10.1200/JCO.2019.37.7_suppl.354 [DOI] [Google Scholar]
- 23.Tannir NM, Forero-Torres A, Ramchandren R, Pal SK, Ansell SM, Infante JR, de Vos S, Hamlin PA, Kim SK, Whiting NC, Gartner EM, Zhao B, Thompson JA (2014) Phase I dose-escalation study of SGN-75 in patients with CD70-positive relapsed/refractory non-Hodgkin lymphoma or metastatic renal cell carcinoma. Investig New Drugs 32:1246–1257. 10.1007/s10637-014-0151-0 [DOI] [PubMed] [Google Scholar]
- 24.Thompson JA, Motzer RJ, Molina AM, Choueiri TK, Heath EI, Redman BG, Sangha RS, Ernst DS, Pili R, Kim SK, Reyno L, Wiseman A, Trave F, Anand B, Morrison K, Donate F, Kollmannsberger CK (2018) Phase I trials of anti-ENPP3 antibody–drug conjugates in advanced refractory renal cell carcinomas. Clin Cancer Res 24:4399–4406. 10.1158/1078-0432.CCR-18-0481 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.von der Maase H, Hansen SW, Roberts JT, Dogliotti L, Oliver T, Moore MJ, Bodrogi I, Albers P, Knuth A, Lippert CM, Kerbrat P, Sanchez Rovira P, Wersall P, Cleall SP, Roychowdhury DF, Tomlin I, Visseren-Grul CM, Conte PF (2000) Gemcitabine and Cisplatin versus methotrexate, vinblastine, doxorubicin, and Cisplatin in advanced or metastatic bladder Cancer: results of a large, randomized, multinational, multicenter, phase III study. J Clin Oncol 18:3068–3077. 10.1200/JCO.2000.18.17.3068 [DOI] [PubMed] [Google Scholar]
- 26.Albiges L, Flippot R, Rioux-Leclercq N, Choueiri TK (2018) Non–clear cell renal cell carcinomas: from shadow to light. J Clin Oncol 36:3624–3631. 10.1200/JCO.2018.79.2531 [DOI] [PubMed] [Google Scholar]
- 27.Haas NB, Lin X, Manola J, Pins M, Liu G, McDermott D, Nanus D, Heath E, Wilding G, Dutcher J (2012) A phase II trial of doxorubicin and gemcitabine in renal cell carcinoma with sarcomatoid features: ECOG 8802. Med Oncol 29:761–767. 10.1007/s12032-011-9829-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Michaelson MD, McKay RR, Werner L, Atkins MB, Van Allen EM, Olivier KM, Song J, Signoretti S, McDermott DF, Choueiri TK (2015) Phase 2 trial of sunitinib and gemcitabine in patients with sarcomatoid and/or poor-risk metastatic renal cell carcinoma: Sunitinib and gemcitabine in RCC. Cancer 121:3435–3443. 10.1002/cncr.29503 [DOI] [PubMed] [Google Scholar]
- 29.Labrijn AF, Janmaat ML, Reichert JM, Parren PWHI (2019) Bispecific antibodies: a mechanistic review of the pipeline. Nat Rev Drug Discov 18:585–608. 10.1038/s41573-019-0028-1 [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.



