throbber
Downloaded from http://aacrjournals.org/clincancerres/article-pdf/12/20/6106/1965492/6106.pdf by guest on 19 July 2022
`
`Cancer Therapy: Clinical
`
`A Phase I Study on Adoptive Immunotherapy Using Gene-Modified
`T Cells for Ovarian Cancer
`Michael H. Kershaw,1,3,4 Jennifer A.Westwood,1,3 Linda L. Parker,1Gang Wang,1,5 Zelig Eshhar,6
`Sharon A. Mavroukakis,1Donald E. White,1John R. Wunderlich,1Silvana Canevari,7 Linda Rogers-Freezer,1
`Clara C. Chen,2 James C. Yang,1Steven A. Rosenberg,1and Patrick Hwu1,5
`
`Abstract Purpose: A phase I study was conducted to assess the safety of adoptive immunotherapy
`using gene-modified autologousTcells for the treatment of metastatic ovarian cancer.
`Experimental Design: T cells with reactivity against the ovarian cancer ^ associated antigen
`a-folate receptor (FR) were generated by genetic modification of autologous T cells with a
`chimeric gene incorporating an anti-FR single-chain antibody linked to the signaling domain of
`the Fc receptor g chain. Patients were assigned to one of two cohorts in the study. Eight patients
`in cohort 1received a dose escalation of Tcells in combination with high-dose interleukin-2, and
`six patients in cohort 2 received dual-specificTcells (reactive with both FR and allogeneic cells)
`followed by immunization with allogeneic peripheral blood mononuclear cells.
`Results: Five patients in cohort 1 experienced some grade 3 to 4 treatment-related toxicity
`that was probably due to interleukin-2 administration, which could be managed using standard
`measures. Patients in cohort 2 experienced relatively mild side effects with grade1to 2 symptoms.
`No reduction in tumor burden was seen in any patient.Tracking 111In-labeled adoptively transferred
`T cells in cohort 1revealed a lack of specific localization of T cells to tumor except in one patient
`where some signal was detected in a peritoneal deposit. PCR analysis showed that gene-
`modified Tcells were present in the circulation in large numbers for the first 2 days after transfer,
`but these quickly declined to be barely detectable 1 month later in most patients. An inhibitory
`factor developed in the serum of three of six patients tested over the period of treatment, which
`significantly reduced the ability of gene-modified Tcells to respond against FR+ tumor cells.
`Conclusions: Large numbers of gene-modified tumor-reactive T cells can be safely given to
`patients, but these cells do not persist in large numbers long term. Future studies need to employ
`strategies to extend T cell persistence. This report is the first to document the use of genetically
`redirected T cells for the treatment of ovarian cancer.
`
`There is increasing interest in the use of immunotherapy for
`the treatment of malignant disease, and some dramatic clinical
`responses have led to intense activity in this field. In particular,
`the success of adoptive immunotherapy as a treatment for
`
`Authors’ Affiliations: 1Surgery Branch, Center for Cancer Research, National
`Cancer Institute; 2Department of Nuclear Medicine, Clinical Center, NIH, Bethesda,
`Maryland; 3Cancer Immunology Research Program, Peter MacCallum Cancer
`Centre; 4Department of Pathology, University of Melbourne, Melbourne, Australia;
`5Department of Melanoma Medical Oncology, The University of Texas M.D.
`Anderson Cancer Center, Houston, Texas; 6Department of Immunology, Weizmann
`Institute of Science, Rehovot, Israel; and 7Istituto Nazionale Tumori, Milan, Italy
`Received 5/16/06; revised 6/26/06; accepted 7/18/06.
`Grant support: The National Health and Medical Research Council (M. Kershaw),
`National Breast Cancer Foundation of Australia (M. Kershaw), Bob Parker Memorial
`Fund (J. Westwood), and Peter MacCallum Cancer Centre Foundation
`(J.Westwood).
`The costs of publication of this article were defrayed in part by the payment of page
`charges. This article must therefore be hereby marked advertisement in accordance
`with 18 U.S.C. Section 1734 solely to indicate this fact.
`Requests for reprints: Patrick Hwu, The University of Texas M.D. Anderson
`Cancer Center, 1515 Holcombe Boulevard, Unit 430, Houston, TX 77030. Phone:
`713-792-2921; Fax: 713-745-1046; E-mail: phwu@mdanderson.org.
`F 2006 American Association for Cancer Research.
`doi:10.1158/1078-0432.CCR-06-1183
`
`melanoma has prompted us to extend this therapy to ovarian
`cancer. Variables important in the application of this therapy
`have been identified in melanoma patients,
`including the
`requirement for tumor antigen-reactive lymphocytes, high-dose
`interleukin-2 (IL-2; ref. 1), and more recently the benefits of
`prior lymphoablation (2).
`However, endogenous tumor-reactive cells cannot be repro-
`ducibly found in ovarian patients. Nevertheless, several tumor-
`associated antigens have been identified for ovarian tumors,
`including Her-2 (3), tumor-associated glycoprotein 72 (4),
`Lewis-Y (5), and a-folate receptor (FR; ref. 6), and monoclonal
`antibodies exist recognizing these antigens. Recombinant genes
`encoding chimeric receptors incorporating antibody specificity
`can be used to genetically modify T cells to endow them with
`activity against tumor cells (7 – 17).
`We have previously reported the generation of FR-specific
`T cells by modification of T cells with a gene encoding a cell
`surface chimeric receptor linking single-chain (scFv) anti-FR to
`the transmembrane and cytoplasmic domains of the Fc receptor
`g chain. The scFv was derived from the MOv18 monoclonal
`antibody (18), and the chimeric gene is referred to as MOv-c.
`We showed that this gene could endow ex vivo transduced
`T cells with the ability to respond against FR+ tumor cells
`
`Clin Cancer Res 2006;12(20) October 15, 2006
`
`6106
`
`www.aacrjournals.org
`
`UPenn Ex. 2038
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`Downloaded from http://aacrjournals.org/clincancerres/article-pdf/12/20/6106/1965492/6106.pdf by guest on 19 July 2022
`
`Adoptive Immunotherapy of Ovarian Cancer
`
`Cancer Institute, and informed consent was obtained from all patients
`before treatment.
`Patient eligibility. Patients had biopsy-proven recurrent, resected
`recurrent, or residual epithelial FR+ ovarian cancer that failed standard
`effective therapy,
`including cisplatin/carboplatin – or paclitaxel-
`containing regimens. Patients ranged in age from 33 to 60 years and
`had clinical Eastern Cooperative Oncology Group performance status of
`0 or 1. Eligibility criteria required serum creatinine levels V1.6 mg/dL
`and bilirubin <2.0 mg/dL. Blood eligibility criteria included hemoglo-
`bin >9.0 g/dL, WBC >3,000/mm3, and platelets >100,000/mm3 and an
`intact immune system as evidenced by a positive reaction to Candida
`albicans skin test, mumps skin test, or tetanus toxoid skin test on a
`standard anergy panel.
`Response assessment. Patients received radiologic evaluation by
`magnetic resonance imaging, computed tomography, or sonography
`immediately before treatment and at completion of therapy. Disease
`response was determined by comparison of pretreatment and post-
`treatment images. In addition, serum CA-125 levels were determined
`following treatment and compared with pretreatment CA-125 levels.
`T cell generation. A detailed description of the generation and
`characterization of T cells used in cohort 1 of the study has been
`published previously (22). Briefly, patient PBMCs derived from
`leukapheresis were stimulated with anti-CD3 (OKT3, Ortho Biotech,
`Raritan, NJ) and human recombinant IL-2 (600 IU/mL; Chiron,
`
`Emeryville, CA). After 3 days of culture, f5  107 to 1  108
`
`lymphocytes were taken and transduced with retroviral vector
`supernatant (Cell Genesys, San Francisco, CA) encoding the chimeric
`MOv-c gene and subsequently selected for gene integration by culture
`in G418.
`For the generation of dual-specific T cells used in cohort 2,
`stimulation of T cells was achieved by coculture of patient PBMCs
`with irradiated (5,000 cGy) allogeneic donor PBMCs from cryopre-
`served apheresis product (mixed lymphocyte reaction). The MHC
`haplotype of allogeneic donors was determined before use, and
`donors that differed in at least four MHC class I alleles from the
`patient were used. Culture medium consisted of AimV medium
`
`serum
`(Invitrogen, Carlsbad, CA) supplemented with 5% human AB
`(Valley Biomedical, Winchester, VA), penicillin (50 units/mL), strepto-
`mycin (50 mg/mL; Bio Whittaker, Walkersville, MD), amphotericin B
`(Fungizone, 1.25 mg/mL; Biofluids, Rockville, MD), L-glutamine
`(2 mmol/L; Mediatech, Herndon, VA), and human recombinant IL-2
`(Proleukin, 300 IU/mL; Chiron). Mixed lymphocyte reaction consisted
`
`of 2  106 patient PBMCs and 1  107 allogeneic stimulator PBMCs in
`
`2 mL AimV per well in 24-well plates. Between 24 and 48 wells were
`cultured per patient for 3 days, at which time transduction was done
`by aspirating 1.5 mL of medium and replacing with 2.0 mL retroviral
`supernatant containing 300 IU/mL IL-2, 10 mmol/L HEPES, and
`8 Ag/mL polybrene (Sigma, St. Louis, MO) followed by covering with
`plastic wrap and centrifugation at 1,000  g for 1 hour at room
`temperature. After overnight culture at 37jC/5% CO2, transduction was
`repeated on the following day, and then medium was replaced after
`another 24 hours. Cells were then resuspended at 1  106/mL in fresh
`medium containing 0.5 mg/mL G418 (Invitrogen) in 175-cm2 flasks for
`5 days before resuspension in media lacking G418.
`Cells were expanded to 2  109 and then restimulated with
`
`allogeneic PBMCs from the same donor to enrich for T cells specific
`for the donor allogeneic haplotype. Restimulation was done by
`
`incubating patient T cells (1  106/mL) and stimulator PBMCs
`(2  106/mL) in 3-liter Fenwall culture bags in AimV + additives and
`IL-2 (no G418). Cell numbers were adjusted to 1  106/mL, and IL-2
`
`was added every 2 days, until sufficient numbers for treatment were
`achieved.
`Cell lines, flow cytometry, and IFN-g secretion assay. Tumor cell
`lines used in assays of T cell function were the FR+ human ovarian
`
`melanoma cell lines Mel 526,
`cancer cell line IGROV-1 (23) and FR
`Mel 624, Mel 888, and Mel 1866 (Surgery Branch, National Cancer
`Institute, Bethesda, MD). Tumor cells were maintained in RPMI
`
`in vitro (19). In addition, adoptive transfer of anti-FR mouse
`T cells could inhibit tumor growth in lung metastases and i.p.
`models of disease in mice (20).
`More recently, we have generated FR-reactive cells from
`populations of T cells with endogenous
`specificity for
`allogeneic antigen. We showed that
`these T cells could
`respond to both tumor and allogeneic antigen and referred
`to these T cells as dual specific (21). The rationale behind the
`generation of dual-specific T cells was to provide a population
`of FR-reactive T cells that could expand in vivo in response
`to allogeneic immunization, which is not possible in response
`to FR alone. Indeed, adoptive transfer of mouse dual-specific
`T cells into mice followed by allogeneic immunization
`resulted in expansion of transferred cells and enhanced inhib-
`ition of s.c. tumor growth without the need for administration
`of IL-2 (21).
`Based on these encouraging results in mice, we initiated a
`two-cohort phase I clinical study in ovarian cancer patients.
`Cohort 1 patients were treated with adoptive transfer of bulk
`peripheral blood – derived T cells gene-modified with the anti-
`FR chimeric receptor in combination with high-dose IL-2.
`Cohort 2 involved the generation of dual-specific T cells from
`autologous peripheral blood mononuclear cells (PBMC) and
`their transfer into patients followed by s.c. immunization with
`allogeneic PBMCs.
`
`Materials and Methods
`
`Treatment regimen. Patients received adoptive transfer of auto-
`logous T cells gene-modified to express a chimeric receptor specific for
`the tumor-associated antigen FR. The study was divided into two
`cohorts, with cohort 1 receiving T cells and high-dose IL-2 and cohort 2
`receiving dual-specific T cells and s.c. immunization with allogeneic
`PBMCs but no IL-2.
`Eight patients were enrolled in cohort 1, each receiving up to three
`cycles of treatment, with each cycle consisting of administration of
`gene-modified T cells and IL-2 (720,000 IU/kg body weight).
`Approximately 4 weeks elapsed between the start of each cycle.
`Following activation, transduction, and G418 selection, T cells were
`expanded in culture, harvested, washed, and resuspended in 100 mL
`of saline and given to patients by i.v. drip over 20 to 30 minutes. The
`first five patients received a dose escalation regimen beginning at
`
`not easily rectified within 24 hours, the patient was eligible to proceed
`
`3  109 transduced T cells. If no grade 3 or 4 toxicity was observed,
`to the next dose level of 1  1010 T cells at the start of the next cycle and
`subsequently to the highest test dose level of 3  1010 to 5  1010 cells
`
`at the start of the third cycle. IL-2 was given i.v. on the day of T cell
`transfer and every 12 hours for up to six doses if tolerated.
`Six patients were enrolled in cohort 2, each receiving up to two cycles
`of treatment, with each cycle consisting of adoptive transfer of gene-
`modified dual-specific T cells followed by immunization with
`allogeneic PBMCs. Eight to 12 weeks elapsed between the start of each
`cycle. Following two in vitro allogeneic stimulations and expansion in
`culture, T cells were given to patients as in cohort 1. Allogeneic
`
`immunization consisted of s.c. injection of f2.0  109 to 4.0  109
`
`allogeneic PBMCs (viable and nonirradiated) from the same donor
`used to stimulate T cells during their generation in vitro. Each dose
`of allogeneic PBMCs was split into four equal parts and injected s.c.
`into separate sites on the lower extremities in 1 mL saline per site.
`Immunization was done 1 day after T cell transfer and again 1 week
`later because multiple allogeneic immunizations had shown better
`effect in mouse studies (21).
`Patient treatment and monitoring procedures were reviewed by the
`Institutional Review Board of the Center for Cancer Research, National
`
`www.aacrjournals.org
`
`6107
`
`Clin Cancer Res 2006;12(20) October 15, 2006
`
`UPenn Ex. 2038
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`Downloaded from http://aacrjournals.org/clincancerres/article-pdf/12/20/6106/1965492/6106.pdf by guest on 19 July 2022
`
`Cancer Therapy: Clinical
`
`supplemented with 10% FCS (Invitrogen), penicillin (50 units/mL),
`streptomycin (50 mg/mL), amphotericin B (1.25 mg/mL), and
`L-glutamine (2 mmol/L). A melanoma-specific T cell
`line used in
`some experiments was derived from tumor-infiltrating lymphocytes of
`a patient at National Cancer Institute and maintained in T cell culture
`medium described above.
`Expression of chimeric MOv-g receptor by transduced T cells
`was determined using flow cytometry following staining with phyco-
`erythrin-conjugated Id18.1, a monoclonal antibody specific for the
`MOv-18 idiotype (24). T cells were stained with phycoerythrin-
`conjugated mouse IgG1 as a control for nonspecific binding.
`Transduced T cells were assessed for their ability to respond against
`the FR antigen by coculture with IGROV-1 ovarian cancer cells. T cells
`
`(1  105) were incubated with IGROV-1 cells (1  105; or FR-negative
`
`control tumor cells) in triplicate wells of 96-well plates. After overnight
`culture, supernatant was taken and assayed for IFN-g using ELISA
`kits according to manufacturer’s instructions (Endogen, Woburn, MA).
`T cells in cohort 2 of the study were also assessed for their ability to
`secrete IFN-g in response to allogeneic stimulator PBMCs freshly
`thawed from cryopreserved stocks. Anti-human CD3 (OKT3) was also
`used to stimulate T cells to gauge their maximal capacity to respond to
`TCR-CD3 engagement. OKT3 was immobilized on 96-well plastic
`plates at 0.5 Ag/well in 100 AL PBS overnight at 4jC.
`In some experiments, a 25% proportion of patient serum was
`included in T cell cocultures to determine possible effects of patient
`serum on T cell function. In some assays, protein G (Amersham
`Biosciences, Piscataway, NJ; 20 AL/mL) was added to serum before
`coculture and incubated for 1 hour at 4jC with gentle rocking to
`deplete patient serum of immunoglobulin.
`Serum FR titer assay. A double-determinant assay was done
`essentially as described (25) using MOv19, a non – cross-reacting
`antibody directed against FR (18), as catcher. Briefly, 96-well flat-
`bottomed maxisorp plates (Nunc, Roskilde, Denmark) were coated
`
`Table 1. Sites of disease and treatment history
`
`Patient
`
`Prior treatment
`
`with 200 AL of MOv19, at 1 Ag/mL in PBS, and incubated overnight
`at 4jC. Plates were washed and blocked for 1 hour with 200 AL/well
`of 0.5% bovine serum albumin in PBS; 100 AL of sample was added to
`wells. A positive control consisted of tissue culture supernatant from
`IGROV-1 cells. Plates were incubated 2 hours and washed, and 100 AL
`of biotinylated MOv18 (0.25 Ag/mL) were added followed by
`incubation at room temperature for 2 hours. Plates were washed,
`and streptavidin-horseradish peroxidase in PBS/0.5% bovine serum
`albumin was added 100 AL/well and incubated for 0.5 hour. Plates were
`washed, and 100 AL/well trimethylbenzidine was added and incubated
`for 5 to 10 minutes, and reaction was stopped with 1 mol/L H2SO4.
`Plates were read on spectrophotometer 450 nm within 0.5 hour of
`stopping reaction. Concentrations of FR in patient sera were expressed
`as dilution until absorbance reached background levels of media alone.
`T cell tracking. A fraction of transduced T cell cultures (17-50%,
`
`1.5  109 to 7.5  109 cells) were radiolabeled with 111In-oxine
`as described previously (26). Briefly, this involved incubation with
`750 ACi 111In-oxine per 1010 cells in 30 to 50 mL PBS for 15 minutes
`with gentle rocking. Labeled cells were then washed and resuspended in
`100 mL of saline containing 5% human serum albumin and 75,000 IU
`IL-2 for i.v. infusion into patients over a period of 10 to 20 minutes.
`Gamma camera images were obtained at intervals for up to 5 days
`where practicable.
`T cell persistence: PCR-ELISA. Patient peripheral blood was ana-
`lyzed for persistence of transduced T cells by detection of the neomycin
`phosphotransferase (neo) gene using a PCR-ELISA DIG Detection kit
`(Roche, Basel, Switzerland), as per the manufacturer’s directions using
`50 pmol forward neo primer (ATTGAACAAGATGGATTGCACGCAG),
`50 pmol reverse neo primer (TCAGAAGAACTCGTCAAGAAGGCG),
`0.25 unit Taq DNA polymerase (Promega, Madison, WI) and 50% by
`volume of patient PBMC lysate. A series of lysed samples of Jurkat-22
`neo cells (containing 1 copy of neo per cell) was prepared as standards,
`consisting of 1% of cells and decreasing in multiples of 2 down to
`
`1
`
`2
`
`3
`
`4
`5
`6
`7
`8
`
`9
`10
`
`11
`
`12
`
`13
`
`14
`
`Hysterectomy, BSO, debulked, Taxol, Carboplatin, Cisplatin,
`bone marrow transplant, etoposide
`THA/BSO, omentectomy, appendectomy, nodectomy,
`Taxol, Cisplatin, Topotecan, Hexamethylmelamine
`Radical hysterectomy, debulked, Carboplatin, Cytoxan,
`Adriamycin, Mitoxantrane, Tamoxifen, etoposide, radiation
`TAH/BSO Taxol, Carboplatin, Doxil, Topotecan, Gemzar
`TAH/BSO, Carboplatin, Cytoxan, Taxol, Topotecan
`Debulked, Carboplatin, Taxol, Doxil
`TAH/BSO, debulked, Carboplatin, Taxol
`TAH/BSO, debulked, Cisplatin, Cytoxan, Carboplatin,
`Taxol, Topotecan, Doxel
`TAH/BSO, debulked, Carboplatin, Taxol, Cytoxan
`TAH/BSO, debulked, Carboplatin, Taxol, Cisplatin, Taxol,
`monoclonal vaccine, Tamoxifen
`TAH/BSO, omentectomy, appendectomy, pancreatic reduction,
`splenectomy, Cytoxan, Cisplatin, vincristine, Hexalen,
`etoposide, Taxol, Carboplatin, Adriamycin, Topotecan, Gemzar
`TAH/BSO, omentectomy, Taxol, Carboplatin, bone marrow
`transplant, Taxane, Doxil, Herceptin, Gemcitabine, Topotecan
`Ovarian cystectomy, hysterectomy, pelvic lymphadenectomy,
`Taxotere, Carboplatin, Topotecan, Doxyl, Gemzar
`TAH/BSO, omentectomy, pelvic and para-aortic lymphadenectomy,
`Taxol, Cisplatin, external beam radiation, Topotecan,
`Thalidomide, etoposide, Hexalen
`
`Metastatic disease status on enrollment in study
`
`Lower abdominal s.c. mass, two inguinal nodules
`
`Retroperitoneal and left cervical lymph nodes
`
`Liver and vaginal cuff
`
`Perihepatic lesion, midabdominal s.c. nodule
`Perihepatic, ascites, sigmoid mass, omental disease
`Pelvic mass, para-aortic adenopathy
`Multiple sites periaortic retroperitoneal adenopathy
`Liver, pericolonic, pelvic lymph node
`
`Liver and rectal muscle mets
`Omentum, peritoneal implants, diaphragm,
`right supraclavicular nodes, pelvis
`Peritoneal implants, left pleural effusion, liver,
`retroperitoneal nodes
`
`Pelvic and mediastinal mets
`
`Omentum, mediastinum
`
`Epigastric intra-abdominal mass, right pelvic mass
`
`NOTE: Patients had advanced ovarian cancer with metastases to various sites. Before enrolling in the current study, total abdominal
`hysterectomy and bilateral salpingo-oophorectomy were done, and most patients had undergone debulking surgery (patients 1-8 enrolled in
`cohort 1 and patients 9-14 in cohort 2).
`Abbreviations: TAH, total abdominal hysterectomy; BSO, bilateral salpingo-oophorectomy.
`
`Clin Cancer Res 2006;12(20) October 15, 2006
`
`6108
`
`www.aacrjournals.org
`
`UPenn Ex. 2038
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`Adoptive Immunotherapy of Ovarian Cancer
`
`Downloaded from http://aacrjournals.org/clincancerres/article-pdf/12/20/6106/1965492/6106.pdf by guest on 19 July 2022
`
`Fig. 1. Growth and phenotype of gene-modified Tcells. A, following allogeneic
`stimulation, 8  107 Tcells were transduced with retroviral vector encoding the
`MOv-g receptor and maintained at 1to 2  106/mL in media containing IL-2.
`Transduced Tcells were restimulated with allogeneic PBMCs on day 21, which
`resulted in further expansion of Tcells. Using this method, large numbers of
`dual-specificTcells could be generated. T cell expansion depicted is for patient 9.
`Representative of all six patients in cohort 2. The phenotype of transduced Tcells
`from cohort 2 of the study was determined with respect to T cell subset markers
`and chimeric receptor expression using specific antibodies and flow cytometry.
`B, although the relative proportions of CD4+ and CD8+ T cells varied between
`patients, the culture was made up predominantly of CD4+ and CD8+ cells as seen in
`the representative plot. C, expression of the chimeric MOv-g receptor was evident
`following staining with anti-idiotype antibody (thick line) compared with isotype
`control antibody (thin line). Representative of all patients.
`
`All T cells were transduced, as indicated by G418 resistance,
`and expressed the chimeric MOv-g receptor, as determined,
`in flow cytometry, by an increase in fluorescence staining in
`presence of Id 18.1, an anti-MOv18 idiotype monoclonal
`antibody compared with staining in presence of isotype control
`antibody (Fig. 1C). Although expressed at
`low level,
`the
`chimeric receptor endowed T cells with the ability to respond
`specifically against FR+ target cells (see next section).
`IFN-g secretion by T cells in response to tumor cells and
`allogeneic stimulator PBMC. T cell cultures from patients in
`cohort 1 were shown to secrete IFN-g specifically in response to
`FR, and this has been previously reported (22). IFN-g levels in
`response to FR+ IGROV-1 cells varied from 1,749 to 28,560
`pg/mL. With respect to patients in cohort 2, an important
`requirement of dual-specific T cells was their ability to respond
`
`0.016%. PCR cycling consisted of 96jC for 6 minutes followed by
`35 cycles of 95jC for 1 minute, 57jC for 1 minute, and 72jC for
`2 minutes. The ELISA was done using 7.5 AL of the PCR product and
`15 pmol/mL probe (Biotin-AGCAAGGTGAGATGACAGGAGAT), with
`hybridization done at 48jC.
`
`Results
`
`Patient characteristics. All patients had been diagnosed with
`metastatic ovarian cancer. Sites of metastases varied between
`patients but
`involved peritoneal disease with lymph node
`involvement (Table 1). Previous treatments received by patients
`before enrollment in the study varied but included surgical
`removal of primary lesion, debulking, and chemotherapy
`(Table 1). Previous therapies ceased at least 2 weeks before
`receiving gene-engineered T cells.
`Characterization of gene-modified T cells used in cohort 1. A
`complete characterization of T cells used in cohort 1 has
`already been described previously (22), but briefly, T cell
`cultures were stimulated with anti-CD3 antibody and were
`shown to expand from 11,000- to 3,000,000-fold. The mean
`time of culture of T cells from patients in cohort 1 was 47 days
`(range, 25-56 days). The T cells secreted IFN-g specifically in
`response to FR and could lyse FR+ tumor cells. Phenotypically,
`the bulk lymphocyte population was composed of both CD4+
`and CD8+ T cells and was shown to consist of a diverse range of
`clones able to secrete a variety of cytokines, including IFN-g,
`IL-10, granulocyte macrophage colony-stimulating factor,
`
`and IL-2, in response to FR. Percentages of CD4+CD8
`T cells
`
`CD8+ T cell percentages
`varied between 1% and 39%, and CD4
`ranged from 47% to 94%.
`In this cohort of the
`Expansion of T cells used in cohort 2.
`study, T cells received an initial stimulation with allogeneic
`stimulator cells and transduction with retroviral vector for a
`culture period of between 21 and 38 days. During this
`stimulation period, T cell expansion from patients varied from
`12- to 325-fold. To further expand T cell numbers and enrich
`for allo-specific T cells, a second stimulation with PBMCs from
`the original PBMC donor was done. This restimulation resulted
`in a further expansion in T cells of f50-fold. A representative
`growth curve for T cells over two stimulations is presented in
`Fig. 1A. The mean time of T cell culture for cohort 2 was
`40.5 days (range, 37-48 days).
`To promote high levels of allo stimulation, stimulator
`PBMCs were HLA typed at MHC class I loci to check for allelic
`differences to patient HLA type. Patient HLA type was
`determined to be largely dissimilar to stimulator HLA, with
`differences in at least four of six alleles.
`Phenotype of T cells in cohort 2. T cells are characterized into
`two major phenotypic subsets, either CD4+ or CD8+, which have
`different fundamental abilities of helper function or cytotoxic
`function, respectively. Because this could affect on the overall
`function of bulk T cell populations and interpretation of clinical
`results, the relative proportions of CD4+ and CD8+ T cells were
`determined for each culture. Following two stimulations with
`allogeneic PBMCs, expression of CD4 and CD8 T cell markers
`was determined using specific monoclonal antibodies and
`
`flow cytometry. Percentages of CD4+CD8
`T cells varied
`
`CD8+ T cell percentages
`between 2% and 82%, and CD4
`
`
`CD8
`cells were present in all
`ranged from 13% to 85%. CD4
`cultures but only as a minor population (2-15%; Fig. 1B).
`
`www.aacrjournals.org
`
`6109
`
`Clin Cancer Res 2006;12(20) October 15, 2006
`
`UPenn Ex. 2038
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`Cancer Therapy: Clinical
`
`Table 2. Anti-FR and anti-allo responses of transduced T cells from patients in cohort 1 and 2 of the study
`
`Cohort 2
`
`Cohort 1, median
`
`Media alone
`
`)
`Melanoma (FR
`IGROV-1 (FR+)
`Allogeneic stimulator PBMCs
`Autologous PBMCs
`OKT3
`
`Pt. 9
`
`36
`35
`1,375
`2,270
`210
`5,784
`
`Pt. 10
`
`Pt. 11
`
`Pt. 12
`
`Pt. 13
`
`Pt. 14
`
`Median
`
`60
`54
`2,960
`1,555
`456
`13,317
`
`34
`28
`8,010
`2,995
`36
`9,620
`
`42
`26
`1295
`5625
`140
`2890
`
`0
`14
`1,340
`>4,320
`39
`>3,760
`
`19
`27
`9,050
`603
`298
`>59,000
`
`35
`28
`2,168
`2,633
`175
`7,702
`
`153
`139
`6,501
`
`7,457
`
`NOTE: T cell reactivity towards the FR tumor antigen and allogeneic stimulator PBMCs was determined by assaying IFN-g secretion (pg/mL)
`using ELISA following overnight incubation of T cells with the targets listed. Plastic-coated anti-CD3 (OKT3) was used as an indicator of maximal
`T cell response. Transduced T cells from all patients were reactive with FR, and T cells from patients in cohort 2 were reactive with allogeneic
`PBMCs. Nontransduced T cells did not respond against IGROV-1, except for patient 14 in whom 548 pg/mL IFN-g was secreted (data not shown).
`
`Downloaded from http://aacrjournals.org/clincancerres/article-pdf/12/20/6106/1965492/6106.pdf by guest on 19 July 2022
`
`to both FR and allogeneic stimulator PBMCs that were to be
`used as immunogen following T cell transfer. IFN-g secretion
`following coculture of T cells with tumor or allogeneic PBMC
`was used as an indicator of T cell response. Although there was
`some variation between patients in IFN-g levels in response to
`
`FR+ IGROV-1 cells (1,295-9,050 pg/mL; Table 2), secretion
`
`melanoma cells
`was always greater than that in response to FR
`(14-54 pg/mL), thereby showing that transduced T cells could
`respond specifically against FR. The specificity of the response
`was also supported by the observed lack of IFN-g secretion
`
`Table 3. Summary of treatment regimen and toxicity for patients receiving gene-modified T cells
`
`Patient
`
`Cycle no.
`
`1
`
`2
`
`3
`
`4
`
`5
`6
`
`7
`8
`
`9
`
`10
`
`11
`12
`13
`14
`
`1
`2
`3
`1
`2
`3
`1
`2
`3
`1
`2
`3
`1
`1
`2
`1
`1
`2
`
`Cycle no.
`
`1
`2
`1
`2
`1
`1
`1
`1
`
`No. T
`cells  10
`3.0
`3.0*
`10.0
`3.0c
`9.0
`47.0c
`3.0
`10.0
`17.5
`3.0
`11.4c
`21.9c
`3.0
`28.57c
`11.0
`22.0c
`44.0c
`43.5
`
`No. T
`cells  10
`46.5
`169.0
`13.17c
`50.0
`4.0
`11.7
`36.7
`9.0
`
`No. IL-2 injections
`
`9
`
`Grades of
`adverse events
`
`Grade 1 and 2
`toxicity events
`
`6
`5
`3
`5
`4
`4
`5
`3
`1
`5
`1
`0
`3
`6
`6
`0
`2
`1
`
`9
`
`No. allo
`immunizations
`
`x
`
`2
`2
`2
`2
`2
`2
`2
`2
`
`1, 2, 3, 4
`2, 3
`2, 3
`1, 2
`
`HB, FAT, NAU, PU, PE
`ED
`ED, LEU, PCD, PU, PE
`PCD, FAT, NAU, BIL, PU
`
`3
`2
`3
`
`3
`
`2
`2
`2
`2, 4
`3
`2
`
`No. allo
`cells  10
`9
`4.55
`6.5
`4.5
`7.0
`7.8
`7.32
`6.72
`7.45
`
`LUO
`
`ASC
`FAT, DIA
`FAT
`RIG, VOM
`
`DYS
`
`Grades of
`adverse events
`
`1, 2
`None
`1, 2
`None
`2
`1
`None
`1, 2
`
`Grade 3 and 4
`toxicity events
`
`LEU, HYP, PCD
`RIG
`HB, HYP
`
`DIA, FAT
`
`HYP, STC
`
`DYS
`
`DYSb
`HYP
`
`Grade 1 and 2
`toxicity events
`
`ISR, URT, DYS
`
`RIG, ISR, NAU
`
`ISR
`ISR
`
`HYP, RIG, FAT,
`ISR, NAU, VOM
`
`Abbreviations: ASC, ascites; HB, hemoglobin; PE, pleural effusion; BIL, bilirubin increased; HYP, hypotension; PU, pulmonary; DIA, diarrhea;
`LEU, leukopenia; RIG, rigors; DYS, dyspnea; LUO, low urine output; STC, sinus tachycardia; ED, Edema; NAU, nausea; URT, urticaria; FAT,
`fatigue; PCD, platelet count decreased; VOM, vomiting; ISR, injection site reaction (allogeneic immunization).
`*No progression to higher dose in this cycle due to grade 4 toxicity in previous cycle.
`cSome of these cells were labeled with 111In for trafficking.
`bOff protocol after one cycle due to dyspnea concerns.
`xDose divided into two to four injections given on day 1 and 8 following MOv-g T cells.
`
`Clin Cancer Res 2006;12(20) October 15, 2006
`
`6110
`
`www.aacrjournals.org
`
`UPenn Ex. 2038
`Miltenyi v. UPenn
`IPR2022-00853
`
`

`

`Adoptive Immunotherapy of Ovarian Cancer
`
`Downloaded from http://aacrjournals.org/clincancerres/article-pdf/12/20/6106/1965492/6106.pdf by guest on 19 July 2022
`
`Fig. 2. Biodistribution of radiolabeled Tcells. Patients received up to 7.5  109
`111In-labeled Tcells, and imaging was done using a gamma camera at intervals
`followingTcell transfer. A, representative image of four transfers that were done
`withTcells that received a single stimulation with OKT3. B, representative image of
`three transfers that were done usingTcells that had received two stimulations with
`OKT3. Radioisotope signal was detected in lungs, liver, and spleen. Radiolabeled
`cells were preferentially retained in lungs of patients that received twice stimulated
`Tcells. C, anteroposterior image of the abdomen of patient 4 at 48 hours after
`receivingTcells in cycle 2 with evidence of Tcell localization to a peritoneal tumor
`(bottom) in addition to localization to liver (top).
`
`has been shown to correlate with response in mice and humans
`(27). To determine if anti-FR – transduced T cells trafficked to
`sites of ovarian cancer metastases, a proportion of T cells in
`cohort 1 were labeled with 111In before adoptive transfer, and
`imaging was done at intervals for up to 5 days later. Three
`patients received radiolabeled T cells during a single cycle of
`therapy, and another two patients received radiolabeled cells
`during two cycles (Table 3). Radiolabeled T cells accumulated
`initially in lung and subsequently in liver and spleen (Fig. 2A).
`Interestingly, T cells persisted longer in the lungs of patients
`who received T cells that had been subjected to relatively
`prolonged culture and restimulation in vitro. Restimulation
`was sometimes done to generate sufficient cell numbers for
`treatment. Restimulation consisted of a second round of
`
`upon coculture of nontransduced T cells with IGROV-1 cells
`(data not shown).
`Transduced T cells from all patients in cohort 2 also
`responded against their specific allogeneic stimulator PBMCs,
`although here again the level of IFN-g secreted varied between
`patients (603-5,625 pg/mL; Table 2). No correlation was
`observed between response against FR and allo PBMCs, with
`the anti-FR response sometimes greater than, and sometimes
`less than, the allo response. However, OKT3-induced IFN-g
`secretion was always greater than that induced by IGROV-1.
`Patient treatment details. Because hi

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