throbber
HHS Public Access
`Author manuscript
`Clin Cancer Res. Author manuscript; available in PMC 2016 July 15.
`
`Published in final edited form as:
`Clin Cancer Res. 2015 July 15; 21(14): 3149–3159. doi:10.1158/1078-0432.CCR-14-1421.
`
`Phase I Hepatic Immunotherapy for Metastases study of intra-
`arterial chimeric antigen receptor modified T cell therapy for
`CEA+ liver metastases
`
`Steven C. Katz1, Rachel A. Burga1, Elise McCormack2, Li Juan Wang3, Wesley Mooring3,
`Gary Point1, Pranay D. Khare4, Mitchell Thorn1, Qiangzhong Ma2, Brian F. Stainken5, Earle
`O. Assanah5, Robin Davies4, N. Joseph Espat1, and Richard P. Junghans2
`
`1Roger Williams Medical Center, Department of Surgery, Providence, RI/Boston University
`School of Medicine, Boston, MA 2Roger Williams Medical Center, Department of Medicine,
`Providence, RI/Boston University School of Medicine, Boston, MA 3Department of Pathology,
`Roger Williams Medical Center, Providence, Rhode Island 4Roger Williams Medical Center, GMP
`Core Facility and Clinical Protocol Office, Providence, RI 5Roger Williams Medical Center,
`Department of Radiology, Providence, RI/Boston University School of Medicine, Boston, MA
`
`Abstract
`
`Purpose—Chimeric antigen receptor modified T cells (CAR-T) have demonstrated encouraging
`results in early-phase clinical trials. Successful adaptation of CAR-T technology for CEA-
`expressing adenocarcinoma liver metastases (LM), a major cause of death in patients with
`gastrointestinal cancers, has yet to be achieved. We sought to test intrahepatic delivery of anti-
`CEA CAR-T through percutaneous hepatic artery infusions (HAI).
`
`Experimental Design—We conducted a phase I trial to test HAI of CAR-T in patients with
`CEA+ LM. Six patients completed the protocol, and 3 received anti-CEA CAR-T HAIs alone in
`dose-escalation fashion (108, 109, and 1010 cells). We treated an additional 3 patients with the
`maximum planned CAR-T HAI dose (1010 cells X 3) along with systemic IL2 support.
`
`Results—Four patients had more than 10 LM and patients received a mean of 2.5 lines of
`conventional systemic therapy prior to enrollment. No patient suffered a grade 3 or 4 adverse
`event related to the CAR-T HAIs. One patient remains alive with stable disease at 23 months
`following CAR-T HAI and 5 patients died of progressive disease. Among the patients in the
`cohort that received systemic IL2 support, CEA levels decreased 37% (range 19–48%) from
`baseline. Biopsies demonstrated an increase in LM necrosis or fibrosis in 4 of 6 patients. Elevated
`serum IFNγ levels correlated with IL2 administration and CEA decreases.
`
`Conclusions—We have demonstrated the safety of anti-CEA CAR-T HAIs with encouraging
`signals of clinical activity in a heavily pre-treated population with large tumor burdens. Further
`clinical testing of CAR-T HAIs for LM is warranted.
`
`Correspondence: Richard P. Junghans, PhD, MD, Department of Medicine, Roger Williams Medical Center, 825 Chalkstone Avenue,
`Providence, RI 02908, Tel: (401) 456-2484, Fax: (401) 456-6708, rjunghans@rwmc.org.
`Conflicts of Interest: The authors have no relevant disclosures.
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`UPenn Ex. 2081
`Miltenyi v. UPenn
`IPR2022-00855
`Page 1
`
`

`

`Katz et al.
`
`INTRODUCTION
`
`Page 2
`
`Liver metastases (LM) are a significant cause of morbidity and mortality in patients with
`gastrointestinal adenocarcinoma. Tumor infiltrating lymphocyte (TIL) studies have revealed
`that host T cell responses to LM are significant correlates of survival (1–5). While those
`who mount effective immune responses to LM tend to have prolonged survival, the vast
`majority of patients succumb to their disease in the context of endogenous immune failure.
`The immunosuppressive nature of the intrahepatic milieu (6–9) may promote the
`development of LM and contribute to aggressive disease biology. Given the favorable
`effects of robust liver TIL responses and the inherent suppressive nature of the intrahepatic
`space, delivery of highly specific T cell products for the treatment of LM is a rational
`approach.
`
`T cells engineered with chimeric antigen receptors (CAR) to enable highly specific tumor
`recognition and killing have gained considerable attention (10–12). Reprogramming T cells
`with CAR genes provides an MHC-independent mechanism for docking with and lysing
`tumor cells. Such modified T cells have been alternatively termed “designer T cells”, “T-
`bodies”, or “CAR-T cells” (13–15). Carcinoembryonic antigen (CEA) is an attractive target
`for CAR-T treatment of adenocarcinoma LM given high levels of CEA expression and the
`ability to measure CEA in serum (16, 17). Upon antigen recognition, anti-CEA CAR-Ts
`proliferate, produce cytokines, and kill target cells (18). Previous clinical studies, which
`evaluated systemic delivery of anti-CEA T cells for metastatic adenocarcinoma,
`demonstrated both promise and dose limiting toxicity (19). To improve the tolerability of
`anti-CEA CAR-Ts for LM in addition to enhancing tumor killing within the liver, we
`studied a regional delivery strategy.
`
`Regional intra-arterial delivery of chemotherapy for LM has been demonstrated to yield
`superior response rates and limited systemic morbidity (20). Prior reports of regionally
`infused adoptive cell therapy products have demonstrated the safety of this approach (21–
`25). We propose that hepatic artery infusion (HAI) of anti-CEA CAR-Ts will limit
`extrahepatic toxicity while optimizing efficacy for treatment of LM. To test the safety and in
`vivo activity of anti-CEA CAR-Ts in patients with LM, we conducted the phase I Hepatic
`Immunotherapy for Metastases (HITM) trial (NCT01373047). We utilized a second-
`generation anti-CEA CAR (18), containing the CD28 co-stimulatory and CD3ζ signaling
`domains. We treated an initial cohort with CAR-T HAI intra-patient dose escalations
`without IL2 support and a second cohort that received fixed CAR-T doses with continuous
`IL2 infusions.
`
`Six patients with LM completed our protocol and we demonstrated that HAIs of anti-CEA
`CAR-Ts were well tolerated with and without IL2 infusion. We also demonstrated in vivo
`activity of CAR-T HAIs in patients with large volume LM refractory to conventional
`treatment. In addition to exploring the safety and efficacy of CAR-T HAIs, we examined
`immunologic correlates of intrahepatic and systemic responses. Our findings support testing
`of CAR-T HAIs for LM in future trials.
`
`Clin Cancer Res. Author manuscript; available in PMC 2016 July 15.
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`UPenn Ex. 2081
`Miltenyi v. UPenn
`IPR2022-00855
`Page 2
`
`

`

`Katz et al.
`
`METHODS
`
`Study design
`
`Page 3
`
`In this phase I study (NCT01373047, RWH 11-335-99) we treated two cohorts of three
`patients with anti-CEA CAR-T HAIs without or with systemic IL2 support (Figure 1).
`Cohort 1 was treated with CAR-T HAIs in intrapatient dose escalation fashion (108, 109, and
`1010 cells) without IL2. Those in the cohort 2 received 3 HAI of 1010 CAR-Ts in addition to
`continuous systemic IL2 infusion at 75,000 U/kg/day via an ambulatory infusion pump.
`
`Eligible patients had measurable unresectable CEA-positive LM or detectable serum CEA
`levels and failed one or more lines of conventional systemic therapy. Minimal extra-hepatic
`disease in the lungs or abdomen was permitted. No commercial sponsor was involved in the
`study. Clinical assessments were scheduled on infusion days, and on days 1, 2, 4, and 7 post-
`infusion. Liver MRI and PET examinations were performed within one month prior to the
`first infusion and then within one month following the third CAR-T HAI. The study
`radiologist (BS) graded responses according to modified RECIST (mRECIST) and immune
`related response criteria (26). Safety evaluation was performed per protocol. Severity of
`adverse events was graded using the National Cancer Institute Common Terminology
`Criteria for Adverse Events version 3.0.
`
`Human CAR-T cell production
`The 2nd generation anti-CEA scfv-CD28/CD3ζ (Tandem) chimeric antigen receptor was
`cloned into the MFG retroviral backbone as previously described (FDA BB IND 10791)
`(18). Briefly, the tandem molecule was generated by fusing the hMN14 sFv-CD8 hinge
`segment of the IgTCR (IgCEA) in the MFG retroviral backbone with a hybrid CD28/CD3ζ
`molecule. The construct was verified by sequencing. The clinical retroviral vector
`supernatant was produced using PG13 cells to generate gibbon ape leukemia virus
`pseudotyped viral particles as described (27). All clinical batches were prepared at Indiana
`University vector production facility (Indianapolis, IN) and stored at −80°C.
`
`Rhode Island Blood Center personnel performed leukapheresis at the Roger Williams
`Medical Center (RWMC, Providence, RI). Anti-CEA CAR-Ts were prepared at the RWMC
`Cell Immunotherapy and Gene Therapy (CITGT) Good Manufacturing Practice (GMP)
`Facility with standard operating procedures (SOPs) for processing, manufacturing,
`expansion, dose harvesting, labeling, storage and distribution. Briefly, patient peripheral
`blood mononuclear cells (PBMCs) were isolated from leukapheresis product using Ficoll
`(Sigma, St; Louis, MO). We then activated PBMCs for 48–72 hours in tissue culture flasks
`(BD Falcon, Franklin Lakes, NJ) containing AIM V media (Life Technologies, Grand
`Island, NY) supplemented with 5% sterile human AB serum (Valley Biomedical,
`Winchester, VA), 50 ng/mL of anti-CD3 monoclonal antibody (OKT3; Ortho Biotech,
`Horsham, PA) and 3000 U/mL of IL2 (Prometheus, San Diego, CA).
`
`Using the spinoculation method (28), 7.2 – 14.4 × 108 T cells were transduced in retronectin
`(Takara Bio Inc, Japan) coated 6-well plates in AIM V media with 5% human AB serum,
`3000 U/ mL of IL2, and protamine sulfate (MP Biomedicals) at low speed centrifugation for
`1 hour at room temperature. Three transductions were carried out over 24-hrs. After
`
`Clin Cancer Res. Author manuscript; available in PMC 2016 July 15.
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`UPenn Ex. 2081
`Miltenyi v. UPenn
`IPR2022-00855
`Page 3
`
`

`

`Katz et al.
`
`Page 4
`
`transduction, cells were washed and incubated for 48–72 hours at 37°C. CAR-Ts were
`further expanded in Lifecell tissue culture bags (Baxter, Deerfield, IL) for 10–14 days.
`CAR-T growth curves and cell viability were examined periodically and cell growth media
`was replaced as required. CAR-Ts were examined by flow cytometry with fluorescently
`labeled antibodies specific for CD3 (UCHT1, Invitrogen, Frederick, MD), CD4 (SK3, BD
`Biosciences, San Jose, CA), CD8 (3B5, Invitrogen), and CAR expression (WI2 antibody,
`Immunomedics, Norris Plains, NJ). The WI2 antibody was prepared as an APC conjugate
`(WI2-APC; Molecular Probes). Flow cytometry was performed on a CyAn (Beckman
`Coulter, Brea, CA) or LSR-II (BD Biosciences, San Jose, CA) machine. In vitro activity of
`patient products was measured by bioluminescence cytotoxicity assay. Luciferase-
`expressing CEA+ tumor cells were mixed with anti-CEA CAR-T at various ratios in 96-well
`round bottom plates and loss of bioluminescence from each well measured (29).
`
`We prepared clinical doses using a Fenwal cell harvester system (Baxter, Deerfield, IL) in
`freezing media containing PlasmaLyte (Baxter), 20% human bovine albumin (Valley
`Biomedicals), 10% DMSO (Bioniche Pharma, Lake Forrest, IL) and IL2. Bacterial and
`fungal cultures were monitored for 14 and 28 days respectively. We performed assays for
`endotoxin with LAL Endotoxin assay kits (Lonza, Walkersville, MD). The clinical dose was
`stored in liquid nitrogen and thawed immediately prior to infusion.
`
`Product delivery
`
`At baseline, a mapping angiogram was performed via a common femoral artery approach.
`The gastroduodenal and right gastric arteries, in addition to other potential sources of
`extrahepatic perfusion, were embolized with microcoils. For CAR-T infusions, the same
`arterial access procedure was carried out and the cells were hand injected via a 60cc syringe
`at a rate of <2cc/second with a total volume of 100cc. Angiography with calibrated contrast
`rate was performed after the first 50cc and at completion of the CAR-T infusion to confirm
`preserved arterial flow. Infusions were delivered into the proper hepatic artery when
`possible. In cases of aberrant hepatic arterial anatomy, where either the right or left hepatic
`artery did not arise from the proper hepatic artery, the dose was split based upon lobar
`volume calculations. In such cases, split doses were delivered separately into the right and
`left hepatic arteries to ensure proportionate CAR-T delivery to both lobes.
`
`Correlative studies
`
`Normal liver and LM core needle (16-gauge) biopsies were obtained under sonographic
`guidance at baseline and at the time of the third CAR-T HAI. Three cores were obtained for
`normal liver and LM, with each confirmed by cytology. For each case, 4–5 μm sections
`were stained with H&E and additional unstained slides were stained with anti-CEA antibody
`(TF 3H8-1, Ventana, Tucson, AZ). All immunohistochemical stains were performed on the
`Ventana Medical System at Our Lady of Fatima Hospital (Providence, RI). All slides were
`reviewed in blinded fashion and graded for necrosis and fibrosis. Fibrosis was scored as
`follows: 0% = grade 0; 5–10% = grade 1; 11–50% = grade 2; >50% = grade 3. Necrosis was
`scored as follows: 0% = grade 0; 0–10% = grade 1; 11–50% = grade 2; >50% = grade 3.
`Flow cytometry was performed on fresh biopsy tissue for CAR-T cells and peripheral blood
`as described above.
`
`Clin Cancer Res. Author manuscript; available in PMC 2016 July 15.
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`UPenn Ex. 2081
`Miltenyi v. UPenn
`IPR2022-00855
`Page 4
`
`

`

`Katz et al.
`
`Page 5
`
`We measured serum IFNγ levels in all patients by ELISA (EBioscience, San Diego, CA).
`Samples were purified with the Purelink DNA Isolation Kit (Life Technologies, Grand
`Island, NY) according to the manufacturer’s instructions. Patient serum was screened for
`anti-CAR antibodies one month after treatment by flow cytometry. We mixed CAR+ or
`CAR- Jurkat cells with 100ul of 1:1 diluted patient serum and then stained with secondary
`goat anti-human immunoglobulin.
`
`CAR DNA was measured from patient whole blood genomic DNA by qPCR performed at
`the Boston University Analytical Core Facility. SYBR Green technology was used and CAR
`positive samples were identified using 100uM 28F2 forward (5′-
`GCAAGCATTACCAGCCCTAT-3′) and zr2 reverse (5′-GTTCTGGCCCTGCTGGTA-3′)
`primers (custom, Sigma Aldrich, St. Louis, MO). Plasmid DNA containing the CAR gene
`was used as a positive control qPCR. Additional primers were used to amplify CD3,
`GAPDH, and RPL13A (Bio-Rad, Hercules, CA). Raw cycle threshold (Ct) values were
`normalized to the average of the two reference genes (RPL13A and GAPDH) and we used
`the DeltaDelta Ct method to analyze the results. Wet-lab validated and MIQE-compliant
`primers were purchased from BioRad (Hercules, CA).
`
`RESULTS
`
`Study design and patient characteristics
`
`We enrolled eight patients with unresectable CEA+ adenocarcinoma LM who progressed on
`an average of 2.5 (range 2–4) lines of conventional systemic therapy (Table 1). Six patients
`completed the protocol (Figure 1A), one patient withdrew due to an unrelated infection prior
`to treatment, and another patient withdrew due to extrahepatic disease progression prior to
`his third CAR-T HAI. Of the patients that completed the protocol, 4 were male and 2 were
`female. Five patients had stage IV colorectal carcinoma and one patient had pancreatobiliary
`ampullary carcinoma. The average age was 57 (range, 51–66). Patients presented with
`substantial disease burdens, with the average size of the largest LM being 8.4 cm (range,
`1.7–14.4) and five patients having more than 10 LM. The mean CEA level upon enrollment
`was 807 ng/ml (range, 2–3265). Five of eight patients had synchronous colorectal LM and
`the mean disease-free interval was 27.3 months (range, 9 to 37) for patients with
`metachronous LM. All further analyses include only the six patients who completed the
`study.
`
`CAR-T cell product assessment
`
`The leukapheresis product from each patient was analyzed by flow cytometry prior to and
`following transduction with anti-CEA CAR construct. For all patients, the mean percentage
`of CD3+ cells following leukapheresis was 55% (range, 12.0–82.0) and increased to 91%
`(range, 72–97) following activation and transduction (Figure 1B). The mean CD4:CD8 ratio
`was 2.4 (range, 1.4–4.7) in the leukapheresis samples and 0.8 (0.2–2.2) in the final products
`(not shown). The transduction efficiency (CAR+) ranged from 10% to 64%, with a mean of
`45% (Figure 1B). Negligible FoxP3 staining was detected among CAR+ T cells prior to
`infusion (not shown). Cells in the final products were 85% viable prior to infusion (range,
`
`Clin Cancer Res. Author manuscript; available in PMC 2016 July 15.
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`UPenn Ex. 2081
`Miltenyi v. UPenn
`IPR2022-00855
`Page 5
`
`

`

`Katz et al.
`
`Page 6
`
`71–95). In vitro cytotoxicity assays confirmed that patient products specifically lysed CEA+
`target cells (Supplemental Figure 1).
`
`CAR-T cell trafficking following regional infusion
`
`Safety data
`
`We obtained CT guided percutaneous biopsies to sample LM and normal liver prior to the
`first CAR-T HAI and at the time of the final HAI. The proportions of CAR-T (CAR+/total
`lymphocyte%) in LM biopsy, normal liver biopsy, and peripheral blood samples were
`determined by flow cytometry. Samples from patient # 7 demonstrated that 0.8% of normal
`liver mononuclear cells were CAR+ following HAI of CAR-T and 6.6% of intratumoral
`mononuclear cells were CAR+ (Figure 1C). We confirmed that that CAR+ cells in the post-
`infusion LM biopsy specimen were CD3+. CAR-T population data in peripheral blood,
`normal liver, and LM are shown for all patients (Supplemental Figure 2, Figure 1D). CAR-
`Ts were more abundant in the LM compared to normal liver in 5 of 6 patients. In patient # 5,
`CAR-T were found to comprise 2.0% of LM mononuclear cells in a sample obtained during
`a microwave ablation procedure 12 weeks following his final CAR-T infusion (not shown).
`In 4 patients, CAR-T were not detectable in peripheral blood but were transiently present in
`P#7 and P#8 at the time of the final infusion, and the levels dropped below detection 3 days
`later. We also performed qPCR on peripheral blood samples taken at day 2 following the
`final infusion; only patient 7 had a measurable increase (1.1-fold) in CAR DNA relative to
`baseline (not shown).
`
`Anti-CAR antibodies were not detected in patient sera one month following CAR-T
`infusion. This was confirmed by screening sera against CAR+ and CAR− target cells and
`staining for anti-human Ig on the CAR+ cells as described in the methods section.
`
`Adverse events (AE) of any grade attributable to any cause were observed in all patients
`who completed the trial (Table 2). The dose in cohort 1 reached the planned maximal HAI
`CAR-T infusion level at 1010 cells. No CAR-T dose reductions were required in cohort 1
`and therefore, all patients in cohort 2 received 3 doses at the 1010 level with IL2 support.
`There were no grade 4 or 5 adverse events. Febrile AEs were observed in 4 patients. Patient
`# 7 experienced grade 3 fever and tachycardia, with a temperature peak of 104°F. The fever
`and tachycardia resolved in patient # 7 after a 50% dose reduction in her systemic IL2
`infusion. Of note, patient # 7 also experienced an increase in her peripheral eosinophil count
`with a peak of 20% and absolute count of 3,740/ml. Given the reported association between
`IL2 infusion and cardiac thrombosis with other features of Loeffler’s syndrome (30), we
`obtained an echocardiogram and electrocardiogram which were normal. The eosinophil
`count returned to normal limits without specific intervention.
`
`Normal liver parenchyma and biliary structures were well preserved following CAR-T
`HAIs. Biopsies from normal liver did not demonstrate increased levels of inflammation or
`fibrosis following CAR-T HAI whether or not systemic IL2 was administered (Figure 2A).
`While all patients experienced transient elevations of alkaline phosphatase (alk phos), total
`bilirubin, and aspartate aminotransferase levels (AST), only patient # 1 experienced grade 3
`elevations and the majority of values did not deviate significantly from baseline levels
`
`Clin Cancer Res. Author manuscript; available in PMC 2016 July 15.
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`UPenn Ex. 2081
`Miltenyi v. UPenn
`IPR2022-00855
`Page 6
`
`

`

`Katz et al.
`
`Page 7
`
`(Figure 2B). Portal pressures and liver synthetic function were not adversely affected by the
`CAR-T HAIs, as reflected by no patient becoming thrombocytopenic (Figure 2C) or
`coagulopathic (Figure 2D).
`
`Clinical activity
`
`At last follow-up, 5 of the 6 heavily pre-treated patients who completed the trial died due to
`disease progression (Table 3). MRI and PET scans were performed in 5 of 6 patients at
`baseline and 2–4 weeks following the third CAR-T HAI. Patient # 8 did not obtain final
`imaging following a return to his native country and ultimately died of disease progression.
`All patients except # 5 were determined to have radiographic disease progression. Patient #
`5 was found to have stable disease by MRI and PET (Supplemental Figure 3A, arrow).
`Patient # 7 developed new lesions and demonstrated an increase in size of some pre-existing
`lesions, while other lesions decreased in size. The lesion in the posterior sector of patient # 7
`that decreased in size on MRI was not visible on PET (Supplemental Figure 3B). More
`medial disease that was decreased in size on MRI was noted to become hypometabolic on
`the post-infusion PET for patient # 7.
`
`As we anticipated limited utility for short follow-up conventional imaging following
`infusion of CAR-T, we measured serum CEA levels at multiple time points following each
`of the three HAIs for each patient. Among the patients in cohort 1, transient decreases in
`serum CEA were demonstrated in two patients following each CAR-T HAI (Figure 3A).
`CEA kinetics were closely paralleled by changes in serum CA19-9 levels (not shown).
`Patient # 4, who presented with hepatobiliary subtype ampullary carcinoma, was the only
`patient without a CEA decrease at any point during the trial and he also had the shortest
`survival time.
`
`The patients in cohort 2 who received systemic IL2 along with anti-CEA CAR-T had more
`favorable CEA responses to treatment. As each of the three patients in cohort 2 required an
`IL2 interruption or dose reduction, which would likely impact CAR-T function, we
`compared CEA levels at baseline with the time point just prior to IL2 dose change. When
`using these time points, all three patients in cohort 2 had decreases in serum CEA
`concentrations (Figure 3A and Table 3). Patients # 7 and # 8 had a 48% and 43% decrease in
`serum CEA concentrations, respectively, prior to IL2 dose interruption or reduction. The
`mean overall survival time for the 6 patients who completed the trial was 30 weeks with a
`median of 15 weeks (range, 8–102). Patient # 5 is alive with disease at 23 months (102
`weeks) following his final CAR-T HAI. Following completion of the HITM trial, patient # 5
`was determined to have stable disease and we performed a microwave ablation of residual
`unresectable tumor (Supplemental Figure 3).
`
`Detecting radiographic responses in heavily pre-treated patients with advanced metastatic
`disease is challenging, and even more so with immunotherapy where intratumoral
`inflammation and edema may minimize the relevance of standard RECIST criteria (26). As
`such, we obtained LM biopsies prior to and following CAR-T HAIs to assess degrees of
`intratumoral necrosis and fibrosis. After review by a blinded pathologist, 4 patients had an
`increase in intratumoral fibrosis and 3 patients were scored as having an increase in necrosis
`
`Clin Cancer Res. Author manuscript; available in PMC 2016 July 15.
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`UPenn Ex. 2081
`Miltenyi v. UPenn
`IPR2022-00855
`Page 7
`
`

`

`Katz et al.
`
`Page 8
`
`within their LM (Figure 3B). An increase in LM fibrosis is demonstrated for patient # 1 and
`a decrease in CEA+ tumor cells by immunohistochemistry for patient # 8 (Figure 3C).
`
`Serum IFNγ concentrations and CEA responses correlate with IL2 administration
`We measured serum IFNγ levels by ELISA at multiple time points. Spikes in IFNγ were
`noted to occur 24–48 hours after doses in all patients, without or with systemic IL2
`(Supplemental Figure 4). Serum CEA changes were compared to peak change in IFNγ for
`each patient (Figure 3D, top). The inverse correlation between peak IFNγ levels and CEA
`change was significant (R=-0.94, p=0.02). All patient HAI CAR-T doses contained a
`quantity of IL2 (600,000 U). The three patients (#6, #7, and # 8) with continuous systemic
`IL2 exposure and largest CAR-T doses had the best CEA responses and the highest mean
`IFNγ levels (P=0.03, Figure 3D, bottom).
`
`DISCUSSION
`
`Our interest in immunotherapy for LM is based upon studies that have demonstrated LM
`patients with robust T cell responses have significantly improved outcomes. However, most
`LM patients fail to mount effective intrahepatic anti-tumor immunity (1). CAR-T
`technology has advanced considerably in recent years and holds tremendous promise (10,
`11) as an immunotherapeutic tool. We chose HAI of CAR-T to minimize immune mediated
`damage to CEA-expressing extrahepatic tissues and based upon the favorable therapeutic
`index of chemotherapy HAIs (20, 31). We established the safety of anti-CEA CAR-T HAIs
`with and without systemic IL2 support, reaching the maximum planned dose of 1010 cells.
`Systemic IL2 support was associated with increased serum IFNγ levels and improved CEA
`responses, at the expense of more severe but manageable adverse events. Although there
`were no radiographic partial or complete responses, 1 of 6 patients had stable disease and is
`alive at 23 months follow-up. Importantly, histologic evidence of increased LM necrosis and
`fibrosis were seen in the majority of subjects following CAR-T HAI.
`
`The safety of CAR-T HAIs is in line with reports from other groups (21, 23, 25, 32) that
`infused non-CAR cellular products into the hepatic circulation. The limited systemic
`exposure of CAR-T in our study subjects likely accounted for the favorable adverse event
`profile. HAI led to preferential accumulation of CAR-T within LM in 5 of 6 HITM patients,
`compared to normal liver and peripheral blood. CAR-Ts were not detected in the peripheral
`blood in 4 of 6 patients and only transiently in patients 7 and 8. Moderate elevations of liver
`function test values were likely related to the CAR-T HAI but did not result in clinically
`significant consequences. Systemic infusion of T cells expressing anti-CEA TCR was
`reported to result in dose-limiting toxicity (19). Similar toxicities have been seen with our
`CAR-T when systemically infused, particularly with IL2 support (RPJ, unpublished data).
`Our continuous ambulatory infusion dose of IL2, 75,000 U/kg/day, is several-fold lower
`than what is given in other protocols (33). Despite the low daily dose of the IL2 in this
`study, 2 patients experienced grade 3 events requiring IL2 dose reductions. We attributed
`these adverse events, including severe pyrexia and colitis, to the IL2 based upon the fact that
`the symptoms resolved promptly upon IL2 dose reduction. We cannot completely exclude
`the possibility that the IL2 activated a small number of systemically circulating anti-CEA
`
`Clin Cancer Res. Author manuscript; available in PMC 2016 July 15.
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`UPenn Ex. 2081
`Miltenyi v. UPenn
`IPR2022-00855
`Page 8
`
`

`

`Katz et al.
`
`Page 9
`
`CAR-T that mediated fever and colitis. Overall, our IL2 infusion strategy was well tolerated
`and the adverse events easily managed by dose reductions.
`
`MRI and PET scans did not demonstrate a response in any patient, while one patient had
`stable disease and is alive more than 23 months following his final CAR-T HAI. Our
`patients were heavily pre-treated with profound disease burdens, with 4 of 6 patients
`presenting with more than 10 LM. Due to rapid disease progression following cessation of
`CAR-T infusions and IL2, we were unable to follow 5 of the 6 patients beyond 2 months,
`when responses to immunotherapy may manifest radiographically (26). We are encouraged
`by the CEA responses in the cohort that received IL2 and the evidence of necrosis and
`fibrosis following CAR-T HAI in several patients. Based on the timing of the biopsies, we
`cannot determine if IL2 alone or IL2 in combination with a higher CAR-T dose in cohort 2
`contributed to histologic findings (34). We also cannot reach definitive conclusions about
`the efficacy of our approach, but speculate that responses would be more favorable in
`patients with lower disease burdens. Interestingly, CEA declines may be inherently
`beneficial given the recently reported pro-angiogenic effects of CEA (35).
`
`To assess trafficking, we performed image-guided core biopsies of pre- and post-infusion
`LM and surrounding liver. We analyzed biopsy specimens for CAR+ cells, and studied the
`cell populations by flow cytometry. CAR+ T cells were present in detectable numbers in
`both tumor and normal liver after HAI, with numbers passing through liver being
`undetectable or only minimally detectable in peripheral blood. Similarly, in a parallel study
`with systemic administration of the same anti-CEA CAR-T, we demonstrated the presence
`of CAR+ cells by immunohistochemistry in normal liver and in tumor after infusion
`(Junghans et al, data not shown), thus confirming trafficking by this independent method.
`
`As to intrahepatic T cell distribution after HAI, Takayama and colleagues found a
`preferential localization in tumor versus normal liver after infusion of radiolabeled tumor
`infiltrating lymphocytes (TILs) (36). Our own flow cytometric analyses of core specimens in
`this study are also consistent with a conclusion of preferential tumor distribution of CAR T
`cells in 4/6 subjects after HAI. However, further directed assessments will be required to
`independently confirm this association on a statistical level.
`
`Performing detailed assessments of CAR-T phenotype and function can be challenging
`when working with a small sub-population of mononuclear cells isolated from core need
`biopsy specimens. Given the technical limitations related to detecting CAR-T in liver
`biopsies following infusion, alternative strategies including molecular imaging with MRI or
`PET scans (37), or radiolabeling as done by Takayama and colleagues (36) above, should
`receive consideration for future trials.
`
`Effective delivery of anti-CEA CAR-T to CEA+ tumor deposits also correlated with
`histologic evidence of tumor killing and serum cytokine surges (38). CEA responses were
`noted in 3 patients, all of whom received systemic IL2. IL2 alone is not expected to affect
`CEA levels, which presumptively implicates this fraction of CAR-T cells that traffic to
`tumor as mediating this effect. Prior work in this lab has shown tumor responses in animals
`
`Clin Cancer Res. Author manuscript; available in PMC 2016 July 15.
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`Author Manuscript
`
`UPenn Ex. 2081
`Miltenyi v. UPenn
`IPR2022-00855
`Page 9
`
`

`

`Katz et al.
`
`Page 10
`
`(34) and in humans (Junghans et al, unpublished results) with CAR-T cells that are
`dependent upon IL2 supplementation.
`
`One potential limitation on the activity of CAR-T in vivo would be the development of anti-
`CAR antibodies that could lead to rapid elimination of CAR+ cells. In the present instance,
`no patient developed an anti-CAR response. In many cases, the CAR includes foreign
`protein with a murine antibody domain that can elicit an immune response. In the current
`anti-CEA CAR, however, a CDR-grafted humanized version of the murine MN14 antibody
`(39) was selected for CAR engineering; such humanized antibodies are known to have much
`reduced immunization potential with only 4% incidence of anti-immunoglobulin responses
`in human clinical trials (40). Thus, the absence of anti-CAR antibody reaction in our patients
`is reassuring but not surprising.
`
`We speculate that improved CAR-T delivery by HAI may decrease the need for
`lymphodepl

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket