Register for free to join our community of investors and share your ideas. You will also get access to streaming quotes, interactive charts, trades, portfolio, live options flow and more tools.
Register for free to join our community of investors and share your ideas. You will also get access to streaming quotes, interactive charts, trades, portfolio, live options flow and more tools.
I think you should apologize for this comment...
(train 5 or 6 lab rats to do the injections, that's about 100/day for each - well within reason.)
Having personal experience with ZFN design and transduction, I am curious about the experimental details of the 5 transductions that were recently done. I would like to practice due diligence within my professional capacity. But I believe that the laboratory scientists should be able to retain in confidence their specialized knowledge and know-how. It is only their innovation and hard work that will activate a chain of profit in the business side. The man who determined the 5 experiments is leading the company in my opinion.
If as investors we could ensure that the laboratory personnel have all the resources they require. That is why the new lab space is a major development, space increases the scale of research operations, and additional hands in the lab can break process bottlenecks. Whenever I get excited about this company I'm thinking about that one "simple" transduction experiment...
Hans E. Bishop
2012 expectations.
Q1- Nothing
Q2-
1. Lonza tech transfer for Contego completed
2. Contego SRAs for other indications executed
Q3- Nothing
Q4-
3. Submit BLA & IND for Accelerated Approval Registration Trial
4. Lonza Complete FDA inspection & manufacturing validation for Contego
In the future I will elaborate on that nature of these events in laymans terms.
Investor presentation .pdf
Several of my previous questions have been answered in this presentation. I encourage you to look at the timeline for the clinical development. This outline should direct our expectations... trading volumes and such...
http://www.genesis-biopharma.com/
http://66.147.244.217/~genesix6/wp//wp-content/uploads/2011/11/Genesis_Biopharma_Overview_November_2011_FINAL.pdf
I have the same sentiments Cancer Hater. To me this company need only coordinate a clinical trial. That is all. And the doctors who have the experience in conducting these trials are involved in the project.
The whole text white paper: Melanoma Til Trial
If this is the published advise of the committee members, it is possible that it reflects the opinions they offer as board members to Genesis on the design of the first Melanoma Til clinical trial.
White Paper on Adoptive Cell Therapy for Cancer with Tumor-Infiltrating Lymphocytes: A Report of the CTEP Subcommittee on Adoptive Cell Therapy
Jeffrey Weber1, Michael Atkins2, Patrick Hwu3, Laszlo Radvanyi3, Mario Sznol4, and Cassian Yee5; on behalf of the Immunotherapy Task Force of the NCI Investigational Drug Steering Committee
+ Author Affiliations
Authors' Affiliations:1Moffitt Cancer Center and Research Institute; 2Dana Farber–Harvard Cancer Center; 3M.D. Anderson Cancer Center; 4Yale University; and 5Fred Hutchinson Cancer Research Center
Corresponding Author:
Jeffrey Weber, Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL 33612. Phone: 813-745-2007; Fax: 813-745-4384. Email: jeffrey.weber@moffitt.org
Abstract
Adoptive T-cell therapy (ACT) using expanded autologous tumor-infiltrating lymphocytes (TIL) and tumor antigen-specific T cell expanded from peripheral blood are complex but powerful immunotherapies directed against metastatic melanoma. A number of nonrandomized clinical trials using TIL combined with high-dose interleukin-2 (IL-2) have consistently found clinical response rates of 50% or more in metastatic melanoma patients accompanied by long progression-free survival. Recent studies have also established practical methods for the expansion of TIL from melanoma tumors with high success rates. These results have set the stage for randomized phase II/III clinical trials to determine whether ACT provides benefit in stage IV melanoma. Here, we provide an overview of the current state-of-the art in T-cell–based therapies for melanoma focusing on ACT using expanded TIL and address some of the key unanswered biological and clinical questions in the field. Different phase II/III randomized clinical trial scenarios comparing the efficacy of TIL therapy to high-dose IL-2 alone are described. Finally, we provide a roadmap describing the critical steps required to test TIL therapy in a randomized multicenter setting. We suggest an approach using centralized cell expansion facilities that will receive specimens and ship expanded TIL infusion products to participating centers to ensure maximal yield and product consistency. If successful, this approach will definitively answer the question of whether ACT can enter mainstream treatment for cancer. Clin Cancer Res; 17(7); 1664–73. ©2011 AACR.
Translational Relevance
Adoptive immunotherapy with tumor-infiltrating lymphocytes (TIL) has been developed for melanoma over the past 20 years, and when used with lymphoid depletion and high-dose interleukin-2 has emerged as a therapy with the potential for long-term survival in patients with large tumor burdens who have failed multiple prior treatments. In this White Paper, we outline the rationale behind the clinical validation of this regimen and suggest a proposal to test whether access to this complex and demanding treatment can be disseminated beyond a few key centers. The widespread adoption of TIL therapy has important implications for patients with melanoma who have failed frontline therapy who might benefit from it, and for its expansion to other histologies.
Current Clinical and Preclinical Data Supporting Further Development of Adoptive Immunotherapy with Tumor-Infiltrating Lymphocytes
In this article our aim is to develop plans to validate the clinical utility of tumor-infiltrating lymphocytes (TIL) adoptive therapy, which is regarded as arguably the most advanced and fully tested adoptive cellular strategy for the treatment of cancer. TIL were first shown to be practical to grow and possess robust antitumor activity in a 1988 paper in which they mediated regression of established poorly immunogenic tumors in murine tumor models together with high-dose interleukin-2 (IL-2; refs. 1–3). This work at the NCI was followed up by the establishment of practical techniques for expanding TIL from patients with metastatic melanoma by using IL-2, followed by a large-scale “rapid expansion protocol” (REP) by using anti-CD3 antibody and IL-2 (4). TIL were employed for adoptive transfer in a small pilot study and shown to induce regression in 11 of 20 patients, a 55% response rate, mostly partial regressions, albeit in patients that were extensively pretreated, and in some cases they were quite sustained over time (4). Median survivals were 11 to 12 months, with modest evidence of long-term survival. However, the long lead time before adoptive transfer required for patients to allow cells to propagate, resulted in a highly selected patient population that also received high-dose IL-2. There was evidence that the adoptively transferred TIL could traffic to tumor deposits (5, 6), and that they were cytolytic, predominantly CD8 T cells that could lyse autologous tumor cells and secrete IFN-? (7–10). The clinical benefit of the adoptive transfer of TIL seemed to correlate with their recognition of tumor and their ability to secrete cytokines. Further refinement of the basic technology by using gene transfer or other technologies confirmed the high response rate in a larger cohort of patients, with the antigen specificity of the TIL and rapid kinetics of growth found to be associated with clinical benefit (11–15). Nonetheless, this technology remained confined to one institution, and the expense involved as well as the initial investment and the high level of selection of patients discouraged its wide dissemination to other centers.
The recognition in murine systems that lymphoid depletion followed by homeostatic T-cell proliferation facilitated a milieu in which a potent vaccination could occur, or an effective adoptive transfer might be able to take advantage of the “space” opened in the lymphoid compartment generated interest in employing adoptive transfer of TIL after lymphodepletion, as opposed to prior efforts in a normal lymphoid milieu. The first reports using cytoxan and fludarabine for nonmyeloablative lymphoid conditioning followed by adoptive transfer of TIL with high-dose IL-2 showed high response rates and a higher proportion of both complete responses (CR) and long-term survivors (16, 17). One significant innovation with that technology was to propagate the TIL in small 24- or 48-well microcultures, then split the growing cultures after several weeks and test individual cultures for their antitumor specificity by using a simple coculture assay with tumor targets with measurement of IFN-? secretion using ELISA as a readout (18). Only rapidly growing microcultures with specificity would be selected for a final rapid expansion with feeder cells and anti-CD3. Although the major pitfall of this approach was that the duration of cell growth from resection was 5 to 6 weeks, resulting in a significant selection for patients without rapidly growing disease, response rates seemed to be high. A significant experience with this methodology allowed an assessment of factors that seemed to correlate with antitumor response and benefit, and consistent with murine findings, memory T cells could be propagated with this technique, and high levels of adoptively transferred CD8 T cells could be found in the peripheral circulation of patients for 6 to 10 months after adoptive transfer, reaching up to 80% of all CD8 T cells in some cases (19–23). The effector cells often had a central memory or effector memory phenotype, and their long-term persistence seemed to be strongly associated with clinical benefit. The presence of IL-7R–expressing memory cells with long telomeres, the rapid pace with which the cells for adoptive transfer were grown, and the persistence of adoptively transferred cells was associated with benefit, yielding the first rigorously derived data on factors that were important for successful adoptive transfer. However, these associations of T-cell memory phenotype with clinical response will need independent verification by other centers performing adoptive T-cell therapy (ACT) with TIL.
More recent innovations have gone beyond the basic TIL technology by taking advantage of the knowledge of and cloning of the clonotypic T cell receptors (TcR) from therapeutically effective TIL cultures to genetically transfer and express the cloned TcR in peripheral blood mononuclear cells (PBMC) that could be quickly and rapidly expanded for adoptive transfer of an oligoclonal, TcR-enriched population of overall effectors (24, 25). This technology has already been used to treat melanoma patients with a modest response rate after additional high-dose IL-2, and was done without lymphoid depletion. This technology can be employed with a TcR for any tumor antigen and theoretically used for any tumor histology, and unpublished reports of antitumor responses in breast, sarcoma, and colon cancer using adoptively transferred PBMC transduced with TcR genes encoding cancer testis and other differentiation antigens have been described.
Subsequent animal work suggested that the more profound the lymphoid depletion, with chemotherapy added to total lymphoid irradiation (TLI), the more likely it was that tumors would regress and animals were cured. This led to further trials in which varying doses of TLI were added to nonmyeloablative treatment with cytoxan at 60 mg/kg on days 1 and 2, and fludarabine at 25 mg/m2 on days 3–7 (26). The largest experience yet published with TIL suggested that highest doses of TLI were associated with the highest response rates of up to 72% and longer median survivals, although small numbers prevented a rigorous comparison of the different lymphoid depletion regimens (27). At least one other center, located in Israel, has published data on their experience with nonmyeloablative lymphoid conditioning followed by TIL and high-dose IL-2, confirming a high response rate and selected long-term survivors (28). Those investigators chose to employ TIL that were not selected after an initial period of culture for tumor cell specificity. The investigators instead just propagated a large selection of TIL from tumor fragments and, as they grew, pooled them together for the REP without regard for their specificity for autologous tumor cells or cell lines, and adoptively transferring the “young TIL” early after culture, at approximately 4 weeks of growth (29). In addition, this clinical trial found selection of TIL cultures based on antitumor reactivity did not result in any significant clinical benefit over “unselected” TIL (29). Recent data from the NCI also support this contention. Although this finding needs to be verified, it suggests that future clinical trials on ACT with TIL will not require selection of cells for further expansion based on antitumor reactivity; a factor that will significantly streamline the process of TIL expansion.
Antigen-Specific CD8+ T Cells
At least 4 groups have isolated and expanded antigen-specific CD8 T cells for adoptive therapy. Mitchell and colleagues (30) used insect cells transduced with HLA-A2 and CD80 to generate cultures of tyrosinase-specific CTL. Up to 5 × 108 T cells (10%–30% of these being tyrosinase-specific) were administered. In the absence of IL-2, transferred T cells were short-lived and only modest clinical responses were observed (30). Conventional APCs (dendritic cells) pulsed with peptides of melanocyte-associated antigens were used by several groups to generate antigen-specific T cells for clinical trials. Using CD8 T-cell clones against gp100, MART1, or tyrosinase antigens, expanded to 1010 cells/m2, the Seattle group showed for the first time that a uniform population of T cells persist in vivo in response to low-dose IL-2, traffic to antigen-positive sites in tumor and skin, elicit autoimmune and tumor-specific responses and mediate clinically relevant responses (5 of 10 patients with stage IV melanoma had stable disease, and an additional 3 patients experiencing some disease regression or a mixed response for up to 11 months; ref. 31). Using unselected, melanoma-reactive cultures, adoptively transferred MART-1–specific T cells, Mackensen and colleagues (32) also achieved favorable clinical responses with 1 CR, 1 partial response (PR), and 1 mixed response of 11 patients with refractory metastatic disease; homing to tumor sites as well as evidence of antigen-loss tumor variants were observed, suggesting an effective epitope-specific immune response.
Conditioning regimens can also modulate the in vivo persistence of adoptively transferred CTL clones. In one study of 14 stage IV melanoma patients, adoptively transferred CD8+ CTL clones infused following a regimen of Decarbazine (DTIC) persisted for more than 30 days postinfusion and produced a response rate of 6 of 14 CR + PR (43%) in patients with metastatic melanoma (33). To define a well-tolerated conditioning regimen, Yee and colleagues evaluated, in sequential fashion, the influence of fludarabine lymphodepletion, using the identical CD8 T-cell clone administered first without and then with conditioning. An increase in serum IL-15 accompanying a 3-fold increase in persistence in vivo was observed among transferred clones following fludarabine compared with no conditioning. However, clinical responses were not substantially improved over previous studies, a result that may be attributable to the rebound increase in the proportion of FoxP3+ regulatory T cells arising after lymphocyte reconstitution (34). The clinical results using antigen-specific clonal T cells as noted earlier are appealing, but no data have been generated to determine whether a clonal or oligoclonal effector cell population would be more clinically beneficial. The more extensive clinical experiences with oligoclonal TIL have established it at this time as the more promising approach.
A somewhat different approach to the use of adoptive therapy was taken by Rapaport and June and colleagues, who have performed several trials in myeloma and other hematopoietic malignancies in which they employ ex vivo anti-CD3/anti-CD28 costimulated autologous T cells after an autologous stem cell transplant in which patients were vaccinated before and after the stem cell transplant. Rapid T-cell reconstitution and delay in T regulatory cell expansion were observed, and a significant proportion of immune responders to the peptides used, with excellent clinical results (35, 36).
In a corollary study, the nonmyeloablative regimen of high-dose cyclophosphamide (4 g/m2) as single agent conditioning prior to the adoptive transfer of antigen-specific CD8 T-cell clones followed by low-dose IL-2 was evaluated and found to be well tolerated with no complications arising from the 7 to 10 day period of leukopenia, yet capable of achieving T-cell frequencies of 1% to 3% more than 12 months after infusion. Four of 6 patients with refractory metastatic melanoma on this study experienced tumor regression including 1 patient with a durable CR, and 4 with PR or mixed response.
Overall, these studies further emphasize the importance of the type and duration of the preconditioning regimens used to transiently deplete lymphocytes and how it alters the “cytokine landscape” and how rapidly specific endogenous T cells reemerge (e.g., Tregs) in the host. All these can greatly affect the persistence and function of the transferred T cells and will need more careful study.
Antigen-Specific CD4+ T-Cell Therapy
CD4 T cells play a central role not only in priming a CD8 response but also in the effector phase of cellular immunity by (i) mediating tumor killing directly against class II+ tumor targets or indirectly against class II- tumors following recognition of cross-presenting class II+ antigen-presenting cells and activation of nonspecific effectors such as macrophages or eosinophils; (ii) supporting the survival of transferred CD8+ CTL via lymphokines and other signals following antigen encounter; and (iii) maintaining CD8 effector function in vivo.
The presence of CD4 T cells in EBV-reactive cell products seems to favor the in vivo persistence of CD8 T cells and induction of antitumor responses in patients with posttransplant lymphoproliferative disease (37), whereas, for melanoma patients, the cocultivated CD4 T cells in TIL cultures (38) and polyclonally expanded antigen-specific CTL may provide a helper response to accompanying CD8 T cells.
The identification of a number of class II–restricted epitopes (e.g., tyrosinase, NY-ESO-1, MAGE-1; ref. 39) afforded the opportunity to evaluate helper CD4 T-cell responses in patients with metastatic melanoma first in vaccine studies and more recently, in a first-in-human clinical trial using antigen-specific CD4 T cells in 9 patients. NY-ESO-1 or tyrosinase-specific Th1-type CD4 T-cell clones were used to treat refractory metastatic melanoma at doses of up to 1010 cells/m2. T-cell frequencies as high as 3% were observed for up to 2 months. Four patients experienced a PR or disease stabilization and 1 patient underwent durable CR of more than 3 years. In some patients, induction of endogenous responses to nontargeted antigens was also observed (i.e., “antigen-spreading”) and may have contributed to a more complete response (40; C. Yee, unpublished data). Antigen spreading, whereby uptake and processing of killed tumor cells by antigen presenting cells can result in cross-presentation of nontargeted antigens and broadening of a focused response, has been observed in previous preclinical and clinical vaccine studies and represents one strategy to circumvent the outgrowth of antigen-loss tumor variants (41–44).
Antigen-Specific T-Cell Therapy of Leukemia
The antileukemic response following a matched allogeneic hematopoietic stem cell transplantation (HSCT) can be attributed in part to donor T-cell responses against recipient minor histocompatability antigens (mHAg). Minor antigens are peptides encoded by polymorphic genes that differ between the donor and recipient tissues and are presented on the cell surface by MHC class I and II molecules. MHAg can elicit CD8+ and CD4+ T-cell responses that initiate and maintain both graft-versus-host-disease (GVHD) and the graft-versus-leukemia (GVL) effect. Much current research is motivated by the hypothesis that selective targeting of mHAg expressed only on recipient normal and malignant hematopoietic cells will mediate an antileukemic effect without triggering or exacerbating GVHD. Since the first mHAgs, H-Y/SMCY (45, 46) and HA-2 (45), were identified in 1995, at least 30 different genes have been shown to encode mHAgs, and the total number is likely to be much higher. Several of the mHAgs that have been identified to date are selectively expressed in hematopoietic cells, suggesting that therapy targeting these antigens could selectively enhance GVL activity without engendering GVHD. In some cases, these antigens have been shown to be expressed on leukemic stem cells; a finding based on studies showing functional eradication of the leukemia-initiating cell in the NOD/SCID transplant model (47, 48). Aberrant expression of some minor antigens has also been seen in solid tumor malignancies (49, 50) and may account for tumor regression observed after allogeneic HSCT (51, 52).
A recent clinical study reports the first-in-human use of adoptively transferred donor-derived T-cell clones targeting tissue-restricted minor antigens for the treatment of patients with relapsing leukemia after allogeneic HSCT. Of 7 patients with recurrent leukemia, 5 achieved a complete, but transient, response to therapy. The most significant toxicity observed was pulmonary toxicity, which was thought to be attributable in at least one instance to shared minor antigen expression in lung epithelium (53). In vivo persistence of transferred T-cell clones was relatively short-lived (21 days), and strategies to extend survival of adoptively transferred cells will be required to prolong the duration of clinical response.
In the autologous setting, T-cell therapy is being developed against a number of leukemia-associated antigens (LAA): WT-1 (54–56), PRAME (57, 58), PR3 (56, 59–61), HNE (62, 63), and several cancer-testis antigens. In spite of the relatively rare and weakly avid endogenous T-cell responses to these self-proteins, the feasibility of generating leukemia-reactive antigen-specific T cells has been established by using peptide-pulsed antigen-presenting cells in vitro (63–65) and for WT-1 it has been translated to early phase clinical studies of adoptive therapy. Recently, the development of tools to positively select antigen-specific CD8+ T cells based on the induction of CD137/4-1BB expression following activation with tumor antigens may facilitate progress in this area.
Adoptive therapy represents an attractive modality for the treatment of leukemic recurrence following allogeneic HSCT if early disease detection and timely generation of mHAg-specific and LAA-specific T cells can be achieved. Concurrent immunosuppressive therapy for treatment of GVHD can impair antitumor immune response, and strategies to render T cells resistant to calcineurin inhibitors (FK6506 and CSA) and steroids are being developed (66, 67).
What Important Scientific Questions Must Be Addressed to Increase the Ability of Investigators to Perform Adoptive Cell Therapy with TIL?
Here, we list the consensus questions regarded as most critical to answer so that the field of adoptive therapy may be advanced.
What is the optimal type of effector cell for adoptive cell therapy of cancer?
Tumor infiltrating lymphocytes
Peripheral T-cell populations and clones
Gene transduced T cells
NK and NKT cells
Antigen presenting cells
Can we expand effector cells to sufficient numbers rapidly enough to make adoptive cell therapy for cancer practical and widespread?
What are the optimal growth conditions for expansion of effector cells?
What are the optimal cytokines/antibodies/chemokines for administration in vivo to patients receiving adoptive cell therapy for cancer?
Which subpopulation of effectors is responsible for mediating regression of tumor and clinical benefit during adoptive cell therapy of cancer?
Can we identify an effector cell phenotype that mediates clinical benefit?
Can we identify an effector cell genotype that mediates clinical benefit?
Can we identify an antigen specificity(ies) that is responsible for clinical benefit?
Is lymphoid depletion/homeostatic proliferation necessary for the success of adoptive cell therapy of cancer?
Can the cell populations that suppress adoptively transferred effectors be defined?
What factors are most importantly involved in resistance to adoptive therapy?
Host-related factors
Tumor-related factors
Ultimately, what will be the best host preconditioning regimen for adoptive therapy?
Cytoxan plus fludarabine
Cytoxan alone
Cytoxan plus fludarabine together with TLI
Targeted lymphocyte depletion (T regulatory Cells, B cells, myeloid-derived suppressor cells).
The answers to these outstanding questions await further clinical trials and preclinical work, such as identifying alternative T-cell expansion strategies (e.g., the use of artificial APC with defined costimulatory molecules). However, as described earlier in the text, we have already made much headway addressing these issues with TIL ACT in melanoma to the point that multicenter phase II and phase III clinical trials testing the efficacy of this therapy by current methodologies can be done to build a firmer foundation on which to further refine the therapy in future clinical investigations.
Can We Define a Proof of Concept Trial for TIL That Will Be a Major Advance in the Field of Adoptive Cell Therapy of Cancer and Push It Forward?
There are 2 types of trials that might be proof of concept trials for adoptive therapy using TIL.
In one design, lesions (at least 2 cm in diameter) would be harvested from patients and TIL expanded. Patients whose TIL expand to a minimum cell number for the subsequent large-scale REP will be randomly allocated to receive their TIL preceded by lymphodepleting cytoxan and fludarabine, followed by high-dose IL-2, or high-dose IL-2 alone at the time that TIL would be ready. Endpoints would be response rate and time to progression, and a crossover would be allowed for the TIL patients to receive high-dose IL-2, and for the IL-2 patients to receive their TIL with cytoxan–fludarabine and IL-2 on progression, negating overall survival as an endpoint. This would answer the pure scientific question of the benefit of TIL versus IL-2 with all patients starting from a baseline of being able to grow TIL, eliminating that factor as a bias.
The alternative design, favored by the Surgery Branch, would involve the same patients, with at least one 2-cm diameter lesion that can be harvested for TIL expansion. But, here, the patients would be randomized at the outset to have the tumor harvest followed by growth of the cells and adoptive transfer, or immediately receive high-dose IL-2 for up to 4 cycles. This would be an intent to treat analysis, because patients who could not wait for the TIL or who did not have TIL that grew, would be counted in the TIL group, stacking the odds against the TIL group. This design would answer the real world question of whether it is truly disadvantageous to wait for TIL growth, and whether in spite of the only 80% success rate of expanding TIL from tumor fragments, and the drop out of some patients during the expansion, it is still superior for the overall group to have TIL expanded and wait for TIL treatment.
Both trials would require approximately 135 patients to have an 80% power to observe a doubling in the response rate and a 50% increase in progression-free survival (PFS), P = 0.05 two-sided.
What Are the Practical Considerations for the Clinical Care of Cancer Patients Using Adoptive Cell Therapy with TIL for Cancer?
The rapid dissemination of adoptive immunotherapy as a research or standard-of-care treatment modality requires the development and/or acquisition of the appropriate infrastructure at an individual institution, and establishment of closely monitored standard operating procedures (SOP) for each step in the process. Some of the issues in establishing an adoptive immunotherapy program within an institution may be similar to those previously encountered and solved for “routine” bone marrow/stem cell transplantation. However, for most forms of adoptive immunotherapy, the additional step of in vitro manipulation, for example, isolation and expansion of antigen-specific T cells, or transfer of genetic material to the cells, could add substantial complexity and cost to the generation of the cell product. Even under the circumstances in which a central (possibly commercial) facility will “produce” the cell product, an institution must have the clinical and regulatory capacity (facilities and personnel) to harvest, ship, receive, perform final testing, and infuse the product safely and effectively. Some of that expertise is already available in those institutions with an active stem cell transplant program.
A very first step in disseminating TIL therapy is the standardization and validation of the processes of tumor harvest, preservation, and shipping, followed by cell preparation at a central facility and shipping back of the prepared product. Each institution that would participate in a TIL trial with central cell growth must show that a system is in place for the reliable shipping of specimens, and the receipt of viable cells for adoptive transfer. Pilot runs of this process for at least 10 times will likely need to be performed at each institution.
Most institutions interested in developing a research program in adoptive immunotherapy will likely require their own laboratory facilities to produce the cell product, although many important research questions can be asked with cells generated by a “standard” central or commercial laboratory, such as expansions of those existing at the National Cancer Institute and at the M.D. Anderson Cancer Center, or commercial facilities. In establishing the internal infrastructure to conduct adoptive immunotherapy trials, the major issues facing the institutional leadership pertain to the type of facility required to generate cell products (and the type of cell products that will be the subject of trials), the number and type of personnel required to adequately staff the facility, the initial cost to build the facility and hire personnel, the cost of maintaining the facility and personnel, the costs of the cell products generated in the facility, the clinical care costs of patients treated with the products, and the ability to ultimately recover the costs of both maintaining the infrastructure and clinical care of the patients. Realistically, recovery of costs may depend on insurance reimbursement (thus the need to conduct trials that show efficacy and possibly cost-effectiveness), grant funding from NIH and other sources, and philanthropy. In addition, decisions to embark on this investment will depend on the clinical and scientific interests and expertise present within the institution. As with bone marrow/stem cell transplantation, demonstration of clear and superior efficacy of adoptive immunotherapy in cancer or other diseases (e.g., in autoimmune disorders), or heightened scientific interest in this area, may generate market-driven forces to establish cell generation facilities within the institution.
It is beyond the scope of this article to outline every requirement of the cell generation process and to provide a careful cost analysis. The U.S. Food and Drug Administration (FDA) Web site provides draft guidelines and points to consider for somatic cell therapy, potency assays, and gene transfer (http://www.fda.gov/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/default.htm). The techniques for isolating and expanding TIL or peripheral blood T cells modified by genetic transfer of tumor-specific T-cell receptors have been published. Modifications that may increase the efficiency of cell expansion in vitro and perhaps reduce overall costs, for example, by use of closed-system bioreactors, have also been explored and published. However, existing gas-permeable cell culture bags have proven adequate for large-scale cell expansion; this approach can easily support a multicenter clinical trial, with new bioreactor technologies envisioned in the future.
It remains difficult to estimate costs to initiate and maintain an adoptive immunotherapy program, because individual institutions may devise different solutions to the creation of a facility, and the costs of cell generation and clinical treatment could vary widely depending on the cell product, number of cells required, type of modification of cells (e.g., gene transfer), inclusion of lymphodepleting preparative regimen and length of hospitalization, and coadministration of other agents with the cells. For example, a modest-sized program to treat 20 patients per year with TIL would likely require a lab PhD director, at least 2 lab technicians, part effort of a regulatory and quality assurance specialist (to manage FDA Investigational New Drugs and assure Good Clinical Practice compliance), and clinical research staff for conduct of the trial. Some have estimated cell generation costs including release testing in the range of $20,000 to $25,000 per patient. Clinical care costs could include tumor biopsy and/or leukopheresis, chemotherapy for lymphodepletion, and the hospitalization including IL-2 and any supportive care medications. Advances in technology could increase or decrease costs. It is reasonable to assume, based on various estimates provided by the authors of this article, several of whom have established adoptive immunotherapy programs within their institutions, that treatment of a patient with T-cell adoptive immunotherapy will cost less than a typical course of treatment with targeted small molecule or antibody signaling antagonists. Given the potential for adoptive immunotherapy to produce durable long-term remissions of disease, it is possible that future trials will show better cost-effectiveness compared with other types of agents.
Other Technical and Logistical Issues to Consider
Reagents
Important reagents that should to be available to the highest GMP grade, in a reliable manner and reasonably priced for effector cell growth would be the following:
Anti-CD3 antibody (OKT3 or equivalent)
Human AB serum
AIM V or other serum-free media
DNAse
Collagenase
Hyaluronidase
IL-2
IL-7
IL-15
Gas permeable culture bags for cell growth
HSA
DMSO
Monitoring
Considerable effort has been put into validation of immunologic monitoring for cancer vaccine and immunotherapy trials by the Cancer Vaccine Consortium (CVC), headed by Dr. Axel Hoos (Bristol-Myers Squibb) and assisted by Dr. Sylvia Janetski (Zellnet Consulting). A series of validation panels have been run to assess the reliability, robustness, sensitivity, specificity, and predictive value of a number of immune assays. The investigators that were part of the proficiency panels determined whether ELISPOT, tetramer, and intracellular cytokine assays could achieve a high level of reliability for use as immune assays in clinical trials (68, 69). Only the ELISPOT assay, a type of assessment also employed by the AIDS Clinical Trials Group, achieved the threshold wherein it was felt to be a practical, reliable, reproducible, and robust assay for clinical use. For groups that would be involved in a clinical trial to validate the growth and adoptive transfer strategy for TIL, central monitoring with a validated protocol for preservation of frozen PBMC perhaps such as those promulgated by the CVC would be useful. The IFN-? ELISPOT assay using autologous tumor and/or HLA matched tumor cell lines would seem to be the choice for immune assays using fresh PBMC, because laboratory SOPs for the assay have been established, with the use of resting cells overnight prior to the assay, the use of pretested human sera to diminish backgrounds, counting methods to detect apoptotic cells, and SOPs for the plate reading as well as for the training of qualified laboratory personnel have been created. A recent CVC Task Force that met to discuss immune monitoring and the use of serum concluded that serum-free media performed as well as serum-media combinations for performance of ELISPOT assays, independent of SOP used, and recovery and viability of cells were unaffected. Additional immune assays that would be important include the use of TcRV-ß spectratyping to detect polarized TcR usage among the adoptively transferred cells, and phenotyping of T cells for memory markers by standard flow cytometry techniques for assessment the kinetics of central memory and effector as well as naive and effector-memory phenotypes. An important assay would detect the persistence of adoptively transferred T cells, a key parameter, but that has yet to be devised. Measurement of relative telomere lengths of infused TIL will also be useful in this regard. For adoptive transfer of clonal tumor antigen-specific T cells, either CD8 or CD4 cells, the use of the antigen-specific ELISPOT assay are important.
A centralized TIL growth facility for multicenter trials
The generation of clinical grade T cells for patient infusion requires a specialized GMP (Good Manufacturing Practices) facility (70) with experienced technical staff familiar with the federal government's GMP guidelines. As these facilities are not commonly available at many academic institutions, the conduct of a multi-institutional randomized study of ACT would be optimally performed with a centralized TIL growth facility. This would also eliminate any possible variables between institutions in the methods and reagents used for T-cell generation. Despite the establishment of detailed SOPs (18), subtle differences in T-cell growth conditions, such as the source of human serum used in the culture media, between institutions may result in changes to the final product. Therefore, having a standardized, central facility to grow T cells for a multicenter trial would enhance consistency between patients. In addition, use of a centralized facility may be the most workable model for the widespread adoption of T-cell therapy should a randomized study show a clear benefit. As the T-cell trials performed to date have all been done in single institutions with their own GMP facilities, the establishment of a centralized facility will require preliminary studies to standardize the processing and shipping of tumors from the operating room to the central facility and the shipping of expanded cells back to the collaborating institutions.
Immune-related response criteria versus RECIST
In some trials of immunotherapeutic reagents, unusual kinetics of antitumor response have been observed, chiefly in trials of the CTLA-4 abrogating antibodies tremelimumab and ipilimumab. They have been observed to fall into 4 patterns; rapid onset of either a CR or a PR by standard Response Evaluation Criteria in Solid Tumors (RECIST) criteria; slow onset of a response preceded by many weeks or even months of stability or of a PR leading slowly to a CR; growth of existing lesions without new disease indicating progression followed by subsequent regression; and development of new lesions in the face of regression or stability of baseline lesions indicating a mixed response, followed by subsequent regression (71, 72). None of these patterns of response have typically been seen in chemotherapy trials, although they are well known to occur in trials of high-dose IL-2 and occasionally other immunotherapies. Many patients may enjoy prolonged periods of time without the need for further therapy, or may be asymptomatic and indeed may not have clinically significant progression of disease. To assess the clinical consequences for patients with these patterns of response and to study their survival, a recent study was conducted in patients receiving the anti–CTLA-4 antibody ipilimumab who had their disease evaluated both by modified WHO criteria and by a new criteria called immune-related response criteria (irRC) designed to capture the benefit of these new patterns of response, and has been recently published (73). The irRC are different in that a patient may be allowed to have development of new disease without progression being declared; they can have 25%, not 20%, increase of disease dimensions, and progression is simply defined by the sum of the longest dimensions of all disease whether defined at baseline or newly developed at the first and subsequent posttreatment evaluations. The use of these modified criteria would have the effect of expanding the category of patients who would be stable or even those who would be considered responders to therapy. When the overall survival outcome for partial and complete responders and stable patients (defined as those with clinical benefit) by modified WHO or the expanded cohort of patients defined by irRC were assessed, the survival curves were virtually identical, whereas the progressors by either criteria had a dismal outcome. The use of irRC added at least another 10% of patients to the category of response or stability that would not have been found by the standard modified WHO criteria. For cancer patients receiving ACT we would argue that the irRC criteria should be adopted, because it is possible that the same unusual pattern of response may be seen, and it is important to increase the likelihood that all patients who may benefit from this complex but novel treatment be captured.
Conclusions
Experience with ACT for melanoma over the past 20 years had provided a strong foundation to test this regimen in a phase II, proof of concept, confirmatory multicenter clinical trial. This phase II trial would be performed to show that this difficult and complex but clinically promising procedure can be carried out as part of a larger effort at multiple institutions to ensure that an adequately powered randomized trial of adoptive therapy is practical. This type of trial would be required to show that there is a survival advantage for a significant cohort of cancer patients for the adoptive therapy strategy compared with some control group, a minimum requirement for taking this procedure forward to a multicenter phase III scenario. The optimal growth schema for TIL with so-called “young” TIL should be employed to ensure that the growth of TIL is practical and simplified. A central facility for the growth of TIL would be a significant strength of such a proposal, and would again ensure that the trial could be carried out in a timely and practical manner. Major obstacles to overcome would be the sterility of the harvested product, the continued QA/QC and quality of the resulting cell product, the assurance that the harvested tumor and final expanded TIL product can be shipped successfully, and that the mature expanded cells could be grown in a timely enough manner to ensure that a profound selection bias would not limit the accrual of, and the interpretation of, such a trial. Given the recent experience with this treatment, we are confident that these obstacles could be overcome.
Disclosure of Potential Conflicts of Interest
No potential conflicts of interest were disclosed.
Acknowledgments
Received August 24, 2010.
Revision received December 29, 2010.
Accepted December 29, 2010.
©2011 American Association for Cancer Research.
Previous Section
References
1.? Yron I, Wood TA Jr, Spiess PJ, Rosenberg SA.. In vitro growth of murine T cells. V. The isolation and growth of lymphoid cells infiltrating syngeneic solid tumors J Immunol 1980;125:238–45. Abstract
2.? Rosenberg SA, Spiess P, Lafreniere R.. A new approach to the adoptive immunotherapy of cancer with tumor-infiltrating lymphocytes Science 1986;233:1318–21. Abstract/FREE Full Text
3.? Spiess PJ, Yang JC, Rosenberg SA.. In vivo antitumor activity of tumor-infiltrating lymphocytes expanded in recombinant interleukin-2 J Natl Cancer Inst 1987;79:1067–75. Medline
4.? Rosenberg SA, Packard BS, Aebersold PM, Solomon D, Topalian SL, Toy ST, et al. Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma A preliminary report. N Engl J Med 1988;319:1676–80.
5.? Fisher B, Packard BS, Read EJ, Carrasquillo JA, Carter CS, Topalian SL, et al. Tumor localization of adoptively transferred indium-111 labeled tumor infiltrating lymphocytes in patients with metastatic melanoma .J Clin Oncol 1989;7:250–61. Abstract
6.? Griffith KD, Read EJ, Carrasquillo JA, Carter CS, Yang JC, Fisher B, et al. In vivo distribution of adoptively transferred indium-111-labeled tumor infiltrating lymphocytes and peripheral blood lymphocytes in patients with metastatic melanoma J Natl Cancer Inst 1989;81:1709–17 Abstract/FREE Full Text
7.? Topalian SL, Solomon D, Rosenberg SA.. Tumor-specific cytolysis by lymphocytes infiltrating human melanomas J Immunol 1989;142:3714–25. Abstract
8.? Barth RJ Jr, Mulé JJ, Spiess PJ, Rosenberg SA.. Interferon gamma and tumor necrosis factor have a role in tumor regressions mediated by murine CD8+ tumor-infiltrating lymphocytes J Exp Med 1991;173:647–58. Abstract/FREE Full Text
9.? Aebersold P, Hyatt C, Johnson S, Hines K, Korcak L, Sanders M, et al. Lysis of autologous melanoma cells by tumor-infiltrating lymphocytes: association with clinical response J Natl Cancer Inst 1991;83:932–7. Abstract/FREE Full Text
10.? Schwartzentruber DJ, Hom SS, Dadmarz R, White DE, Yannelli JR, Steinberg SM, et al. In vitro predictors of therapeutic response in melanoma patients receiving tumor-infiltrating lymphocytes and interleukin-2 J Clin Oncol 1994;12:1475–83. Abstract
11.? Rosenberg SA, Aebersold P, Cornetta K, Kasid A, Morgan RA, Moen R, et al. Gene transfer into humans–immunotherapy of patients with advanced melanoma, using tumor-infiltrating lymphocytes modified by retroviral gene transduction N Engl J Med 1990;323:570–8. Medline
12.? Rosenberg SA, Yannelli JR, Yang JC, Topalian SL, Schwartzentruber DJ, Weber JS, et al. Treatment of patients with metastatic melanoma with autologous tumor-infiltrating lymphocytes and interleukin 2 J Natl Cancer Inst 1994;86:1159–66. Abstract/FREE Full Text
13.? Kawakami Y, Eliyahu S, Jennings C, Sakaguchi K, Kang X, Southwood S, et al. Recognition of multiple epitopes in the human melanoma antigen gp100 by tumor-infiltrating T lymphocytes associated with in vivo tumor regression J Immunol 1995;154:3961–8. Abstract
14.? Yannelli JR, Hyatt C, McConnell S, Hines K, Jacknin L, Parker L, et al. Growth of tumor-infiltrating lymphocytes from human solid cancers: summary of a 5-year experience Int J Cancer 1996;65:413–21. CrossRefMedline
15.? Kawakami Y, Dang N, Wang X, Tupesis J, Robbins PF, Wang RF, et al. Recognition of shared melanoma antigens in association with major HLA-A alleles by tumor infiltrating T lymphocytes from 123 patients with melanoma J Immunother 2000;23:17–27. CrossRefMedline
16.? Dudley ME, Wunderlich J, Nishimura MI, Yu D, Yang JC, Topalian SL, et al. Adoptive transfer of cloned melanoma-reactive T lymphocytes for the treatment of patients with metastatic melanoma J Immunother 2001;24:363–73. CrossRefMedline
17.? Dudley ME, Wunderlich JR, Robbins PF, Yang JC, Hwu P, Schwartzentruber DJ, et al. Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes Science 2002; 298::850–4 Abstract/FREE Full Text
18.? Dudley ME, Wunderlich JR, Shelton TE, Even J, Rosenberg SA.. Generation of tumor-infiltrating lymphocyte cultures for use in adoptive transfer therapy for melanoma patients J Immunother 2003;26:332–42. CrossRefMedline
19.? Powell DJ Jr, Dudley ME, Robbins PF, Rosenberg SA.. Transition of late-stage effector T cells to CD27 +CD28+ tumor-reactive effector memory T cells in humans after adoptive cell transfer therapy Blood 2005;105:241–50. Abstract/FREE Full Text
20.? Rosenberg SA, Dudley ME.. Cancer regression in patients with metastatic melanoma after the transfer of autologous antitumor lymphocytes .Proc Natl Acad Sci U S A 2004;101:14639–45. Abstract/FREE Full Text
21.? Robbins PF, Dudley ME, Wunderlich J, El-Gamil M, Li YF, Zhou J, et al. Cutting edge: persistence of transferred lymphocyte clonotypes correlates with cancer regression in patients receiving cell transfer therapy J Immunol 2004;173:7125–30. Abstract/FREE Full Text
22.? Zhou J, Shen X, Huang J, Hodes RJ, Rosenberg SA, Robbins PF.. Telomere length of transferred lymphocytes correlates with in vivo persistence and tumor regression in melanoma patients receiving cell transfer therapy J Immunol 2005;175:7046–52. Abstract/FREE Full Text
23.? Shen X, Zhou J, Hathcock KS, Robbins P, Powell DJ Jr, Rosenberg SA, et al. Persistence of tumor infiltrating lymphocytes in adoptive immunotherapy correlates with telomere length J Immunother 2007;30:123–9. CrossRefMedline
24.? Johnson LA, Heemskerk B, Powell DJ Jr, Cohen CJ, Morgan RA, Dudley ME, et al. Gene transfer of tumor-reactive TCR confers both high avidity and tumor reactivity to nonreactive peripheral blood mononuclear cells and tumor-infiltrating lymphocytes J Immunol 2006;177:6548–59. Abstract/FREE Full Text
25.? Yang S, Cohen CJ, Peng PD, Zhao Y, Cassard L, Yu Z, et al. Development of optimal bicistronic lentiviral vectors facilitates high-level TCR gene expression and robust tumor cell recognition Gene Ther 2008;15:1411–23. CrossRefMedline
26.? Dudley ME, Wunderlich JR, Yang JC, Sherry RM, Topalian SL, Restifo NP, et al. Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma J Clin Oncol 2005;23:2346–57. Abstract/FREE Full Text
27.? Dudley ME, Yang JC, Sherry R, Hughes MS, Royal R, Kammula U, et al. Adoptive cell therapy for patients with metastatic melanoma: evaluation of intensive myeloablative chemoradiation preparative regimens J Clin Oncol 2008;26:5233–9. Abstract/FREE Full Text
28.? Besser MJ, Shapira-Frommer R, Treves AJ, Zippel D, Itzhaki O, Hershkovitz L, et al. Clinical responses in a phase ii study using adoptive transfer of short-term cultured tumor infiltration lymphocytes in metastatic melanoma patients Clin Cancer Res 2010; 16: 2646–55. Abstract/FREE Full Text
29.? Tran KQ, Zhou J, Durflinger KH, Langhan MM, Shelton TE, Wunderlich JR, et al. Minimally cultured tumor-infiltrating lymphocytes display optimal characteristics for adoptive cell therapy J Immunother 2008;31:742–51. CrossRefMedline
30.? Mitchell MS, Darrah D, Yeung D, Halpern S, Wallace A, Voland J, et al. Phase I trial of adoptive immunotherapy with cytolytic T lymphocytes immunized against a tyrosinase epitope JClin Oncol 2002;20:1075–86 Abstract/FREE Full Text
31.? Yee C, Thompson JA, Byrd D, Riddell SR, Roche P, Celis E, et al. Adoptive T cell therapy using antigen-specific CD8+ T cell clones for the treatment of patients with metastatic melanoma: in vivo persistence, migration, and antitumor effect of transferred T cells .Proc Natl Acad Sci U S A 2002;99:16168–73. Abstract/FREE Full Text
32.? Mackensen A, Meidenbauer N, Vogl S, Laumer M, Berger J, Andreesen R, et al. ( Phase I study of adoptive T-cell therapy using antigen-specific CD8+ T cells for the treatment of patients with metastatic melanoma J Clin Oncol 2006;24:5060–9. Abstract/FREE Full Text
33.? Kammari A, Labarruewre N, Vignard V, Nguyen JM, Pandolfino MC, Knol AC, et al. Treatment of metastatic melanoma with autologous Melan-A/MART-1 cytotoxic T lymphocyte clones J Invest Derm 2009;129:2835–42. CrossRefMedline
34.? Wallen H, Thompson JA, Reilly JZ, Rodmyre RM, Cao J, Yee C.. Fludarabine modulates immune response and extends in vivo survival of adoptively transferred CD8 T cells in patients with metastatic melanoma PLoS One 2009;4:e4749. CrossRefMedline
35.? Rapoport AP, Aqui NA, Stadtmauer EA, Vogl DT, Fang HB, Cai L, et al. Combination immunotherapy using adoptive T-cell transfer and tumor antigen vaccination on the basis of hTERT and survivin after ASCT for myeloma. Blood 2011;; 117: : 788–97. Abstract/FREE Full Text
36.? Rapoport AP, Stadtmauer EA, Aqui N, Vogl D, Chew A, Fang HB, et al. Rapid immune recovery and graft-versus-host disease-like engraftment syndrome following adoptive transfer of Costimulated autologous T cells .Clin Cancer Res 2009;15:4499–507. Abstract/FREE Full Text
37.? Hanley PJ, Cruz CR, Savoldo B, Leen AM, Stanojevic M, Khalil M, et al. Functionally active virus-specific T cells that target CMV, adenovirus, and EBV can be expanded from naive T-cell populations in cord blood and will target a range of viral epitopes Blood 2009;114:1958–67. Abstract/FREE Full Text
38.? Sun JC, Bevan MJ.. Defective CD8 T cell memory following acute infection without CD4 T cell help Science 2003;300:339–42. Abstract/FREE Full Text
39.? Topalian SL, Gonzales MI, Parkhurst M, Li YF, Southwood S, Sette A, et al. Melanoma-specific CD4+ T cells recognize nonmutated HLA-DR-restricted tyrosinase epitopes J Exp Med 1996;183:1965–71. Abstract/FREE Full Text
40.? Hunder NN, Wallen H, Cao J, Hendricks DW, Reilly JZ, Rodmyre R, et al. Treatment of metastatic melanoma with autologous CD4+ T cells against NY-ESO-1 N Engl J Med 2008;358:2698–703. CrossRefMedline
41.? Boon T, Van Pel A.. Teratocarcinoma cell variants rejected by syngeneic mice: protection of mice immunized with these variants against other variants and against the original malignant cell line Proc Natl Acad Sci U S A 1978;75:1519–23. Abstract/FREE Full Text
42.? Lally KM, Mocellin S, Ohnmacht GA, Nielsen MB, Bettinotti M, Panelli MC, et al. Unmasking cryptic epitopes after loss of immunodominant tumor antigen expression through epitope spreading Int J Cancer 2001;93:841–7. CrossRefMedline
43.? el-Shami K, Tirosh B, Bar-Haim E, Carmon L, Vadai E, Fridkin M, et al. MHC class I-restricted epitope spreading in the context of tumor rejection following vaccination with a single immunodominant CTL epitope Eur J Immunol 1999;29:3295–301. CrossRefMedline
44.? Brossart P, Wirths S, Stuhler G, Reichardt VL, Kanz L, Brugger W, et al. ( Induction of cytotoxic T-lymphocyte responses in vivo after vaccinations with peptide-pulsed dendritic cells Blood 2000;96:3102–8. Abstract/FREE Full Text
45.? den Haan JM, Sherman NE, Blokland E, Huczko E, Koning F, Drijfhout JW, et al. Identification of a graft versus host disease-associated human minor histocompatibility antigen Science 1995;268:1476–80. Abstract/FREE Full Text
46.? Rosinski KV, Fujii N, Mito JK, Koo KK, Xuereb SM, Sala-Torra O, et al. DDX3Y encodes a class I MHC-restricted H-Y antigen that is expressed in leukemic stem cells Blood 2008;111:4817–26. Abstract/FREE Full Text
47.? Bonnet D, Warren EH, Greenberg PD, Dick JE, Riddell SR.. CD8(+) minor histocompatibility antigen-specific cytotoxic T lymphocyte clones eliminate human acute myeloid leukemia stem cells Proc Natl Acad Sci U S A 1999;96:8639–44. Abstract/FREE Full Text
48.? Miyazaki M, Akatsuka Y, Nishida T, Fujii N, Hiraki A, Ikeda K, et al. Potential limitations in using minor histocompatibility antigen-specific cytotoxic T cells for targeting solid tumor cells Clin Immunol 2003;107:198–201. CrossRefMedline
49.? Klein CA, Wilke M, Pool J, Vermeulen C, Blokland E, Burghart E, et al. The hematopoietic system-specific minor histocompatibility antigen HA-1 shows aberrant expression in epithelial cancer cells J Exp Med 2002;196:359–68. Abstract/FREE Full Text
50.? Tykodi SS, Fujii N, Vigneron N, Lu SM, Mito JK, Miranda MX, et al. C19orf48 encodes a minor histocompatibility antigen recognized by CD8+ cytotoxic T cells from renal cell carcinoma patients Clin Cancer Res 2008;14:5260–9. Abstract/FREE Full Text
51.? Spierings E.. Minor histocompatibility antigens: targets for tumour therapy and transplant tolerance Int J Immunogenet 2008;35:363–6. CrossRefMedline
52. Warren EH, Fujii N, Akatsuka Y, Chaney CN, Mito JK, Loeb KR, et al. Therapy of relapsed leukemia after allogeneic hematopoietic cell transplantation with T cells specific for minor histocompatibility antigens Blood 2010;115:3869–78. Abstract/FREE Full Text
53.? Bergmann L, Miething C, Maurer U, Brieger J, Karakas T, Weidmann E, et al. High levels of Wilms' tumor gene (wt1) mRNA in acute myeloid leukemias are associated with a worse long-term outcome Blood 1997;90:1217–25. Abstract/FREE Full Text
54.? Ohminami H, Yasukawa M, Fujita S.. HLA class I-restricted lysis of leukemia cells by a CD8(+) cytotoxic T-lymphocyte clone specific for WT1 peptide Blood 2000;95:286–93. Abstract/FREE Full Text
55.? Scheibenbogen C, Letsch A, Thiel E, Schmittel A, Mailaender V, Baerwolf S, et al. CD8 T-cell responses to Wilms tumor gene product WT1 and proteinase 3 in patients with acute myeloid leukemia Blood 2002;100:2132–7. Abstract/FREE Full Text
56.? van Baren N, Chambost H, Ferrant A, Michaux L, Ikeda H, Millard I, et al. PRAME, a gene encoding an antigen recognized on a human melanoma by cytolytic T cells, is expressed in acute leukaemia cells Br J Haematol 1998;102:1376–9. CrossRefMedline
57.? Kessler JH, Beekman NJ, Bres-Vloemans SA, Verdijk P, van Veelen PA, Kloosterman-Joosten AM, et al. Efficient identification of novel HLA-A(*)0201-presented cytotoxic T lymphocyte epitopes in the widely expressed tumor antigen PRAME by proteasome-mediated digestion analysis J Exp Med 2001;193:73–88. Abstract/FREE Full Text
58.? Molldrem JJ, Clave E, Jiang YZ, Mavroudis D, Raptis A, Hensel N, et al. Cytotoxic T lymphocytes specific for a nonpolymorphic proteinase 3 peptide preferentially inhibit chronic myeloid leukemia colony-forming units Blood 1997;90:2529–34. Abstract/FREE Full Text
59.? Rezvani K, Grube M, Brenchley JM, Sconocchia G, Fujiwara H, Price DA, et al. Functional leukemia-associated antigen-specific memory CD8 +T cells exist in healthy individuals and in patients with chronic myelogenous leukemia before and after stem cell transplantation Blood 2003;102:2892–900. Abstract/FREE Full Text
60.? Yong AS, Rezvani K, Savani BN, Eniafe R, Mielke S, Goldman JM, et al. High PR3 or ELA2 expression by CD34+ cells in advanced-phase chronic myeloid leukemia is associated with improved outcome following allogeneic stem cell transplantation and may improve PR1 peptide-driven graft-versus-leukemia effects Blood 2007;110:770–5. Abstract/FREE Full Text
61.? Fouret P, du Bois RM, Bernaudin JF, Takahashi H, Ferrans VJ, Crystal RG, et al. ( Expression of the neutrophil elastase gene during human bone marrow cell differentiation J Exp Med 1989;169:833–45. Abstract/FREE Full Text
62.? Fujiwara H, El Ouriaghli F, Grube M, Price DA, Rezvani K, Gostick E, et al. Identification and in vitro expansion of CD4+ and CD8+ T cells specific for human neutrophil elastase Blood 2004;103:3076–83. Abstract/FREE Full Text
63.? Weber G, Karbach J, Kuci S, Kreyenberg H, Willasch A, Koscielniak E, et al. WT1 peptide-specific T cells generated from peripheral blood of healthy donors: possible implications for adoptive immunotherapy after allogeneic stem cell transplantation Leukemia 2009;23:1634–42. CrossRefMedline
64.? Ho WY, Nguyen HN, Wolfl M, Kuball J, Greenberg PD.. In vitro methods for generating CD8+ T-cell clones for immunotherapy from the naive repertoire J Immunol Methods 2006;310:40–52. CrossRefMedline
65.? Brewin J, Mancao C, Straathof K, Karlsson H, Samarasinghe S, Amrolia PJ, et al. Generation of EBV-specific cytotoxic T cells that are resistant to calcineurin inhibitors for the treatment of posttransplantation lymphoproliferative disease Blood 2009;114:4792–803. Abstract/FREE Full Text
66.? De Angelis B, Dotti G, Quintarelli C, Huye LE, Zhang L, Zhang M, et al. Generation of Epstein-Barr irus-specific cytotoxic T lymphocytes resistant to the immunosuppressive drug tacrolimus (FK506) Blood 2009;114:4784–91. Abstract/FREE Full Text
67.? Berger C, Turtle CJ, Jensen MC, Riddell SR.. Adoptive transfer of virus-specific and tumor-specific T cell immunity Curr Opin Immunol 2009;21:224–32. CrossRefMedline
68.? Janetzki S, Panagea K, Ben-Porat L, Boyer J, Britten CM, Clay TM, et al. Results and harmonization guidelines from two large-scale international ELISPOT proficiency panels conducted by the Cancer Vaccine Consortium (CVS/SVI) Cancer Immunol Immunother 2008;57:303–15. CrossRefMedline
69.? Janetzki S, Price L, Britten CM, van der Burg SH, Caterini J, Currier JR, et al. Performance of serum-supplemented and serum free media in IFNgamma ELISPOT assays for human T cells Cancer Immunol Immunother 2010;59:609–18. CrossRefMedline
70.? Burger SR.. Current regulatory issues in cell and tissue therapy Cytotherapy 2003;5:289–98. CrossRefMedline
71.? Weber JS, O'Day S, Urba W, Powderly J, Nichol G, Yellin M, et al. Phase I/II study of ipilimumab for patients with metastatic melanoma J Clin Oncol 2008;26:5950–6. Abstract/FREE Full Text
72.? Saenger YM, Wolchok JD.. The heterogeneity of the kinetics of response to ipilimumab in metastatic melanoma: patient cases Cancer Immun 2008;8:1. Medline
73.? Wolchok JD, Hoos A, O'Day S, Weber JS, Hamid O, Lebbé C, et al. Guidelines for the evaluation of immune therapy activity in solid tumors: immune related response criteria Clin Can Res 2009;15:7412–20.
Transparency- Sci Adv Board Describes Clinical Trial Options
This post contains the abstract of a report published by several members of the Scientific Advisory Board detailing the approach they recommend for the upcoming clinical trial. Granted this is just the abstract and I haven't got access to the full white paper yet. I hope enclosed will be the best guidance from "Genesis" as to the plans for the trial, and the strategy for gaining FDA approval. In an upcoming post I will included the link to the full report and some research into the other indications (cancer types) that Genesis suggests they will target in Phase 1 trials after melanoma.
Yet again, I believe this is further evidence of the efforts of the board members, and that this in my humble opinion suggest that they are playing a significant role in the direction of the research and clinical trial efforts of this outfit as they move forward to FDA approval of TIL therapy. This is as it should be. It seem like the clinical superteam is assembled to bring this therapy to market...The incentive is there, patients want this therapy because it works, I'll try to bring more information as I find it to this board and keep up where Genesis is leaving off in details of the treatment, and the rich foundation upon which it is based.
Clin Cancer Res. 2011 Apr 1;17(7):1664-73. Epub 2011 Feb 15.
White paper on adoptive cell therapy for cancer with tumor-infiltrating lymphocytes: a report of the CTEP subcommittee on adoptive cell therapy.
Weber J, Atkins M, Hwu P, Radvanyi L, Sznol M, Yee C;
Immunotherapy Task Force of the NCI Investigational Drug Steering Committee.
Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA. jeffrey.weber@moffitt.org
Abstract:
Adoptive T-cell therapy (ACT) using expanded autologous tumor-infiltrating lymphocytes (TIL) and tumor antigen-specific T cell expanded from peripheral blood are complex but powerful immunotherapies directed against metastatic melanoma. A number of nonrandomized clinical trials using TIL combined with high-dose interleukin-2 (IL-2) have consistently found clinical response rates of 50% or more in metastatic melanoma patients accompanied by long progression-free survival. Recent studies have also established practical methods for the expansion of TIL from melanoma tumors with high success rates. These results have set the stage for randomized phase II/III clinical trials to determine whether ACT provides benefit in stage IV melanoma. Here, we provide an overview of the current state-of-the art in T-cell-based therapies for melanoma focusing on ACT using expanded TIL and address some of the key unanswered biological and clinical questions in the field. Different phase II/III randomized clinical trial scenarios comparing the efficacy of TIL therapy to high-dose IL-2 alone are described. Finally, we provide a roadmap describing the critical steps required to test TIL therapy in a randomized multicenter setting. We suggest an approach using centralized cell expansion facilities that will receive specimens and ship expanded TIL infusion products to participating centers to ensure maximal yield and product consistency. If successful, this approach will definitively answer the question of whether ACT can enter mainstream treatment for cancer.
PMID: 21325070 [PubMed - indexed for MEDLINE]
Understanding what it is like to Cure Cancer.
It has been done at the NIH and the Rosenberg Lab this has been achieved by two members of the scientific advisory board. Rosenberg, and Dudley. I think that this is like walking on the moon for a doctor. The advances they made to develop this treatment are implicit in the fact that this therapy is not just a "pill".
" The actuarial 3- and 5-year survival rates for the entire group were 36% and 29%, respectively, but for the 20 complete responders were 100% and 93%. "
This report is a compiled analysis of three clinical trial, all slightly different, but with the same core technology as Contego. No matter which version, or level of variation adopted by Genesis for the first clinical trial, this report suggests that the variation permissible for success is within the limits of the three trials at the NCI.
Informed investors will want to see the similarity and differences in the clinical trial design from that in practice at the NCI and MD Anderson Cancer Center. Alterations made for cost reduction should be prudent to ensure a response greater than the standard of care. This will be the topic of a future post.
Clin Cancer Res. 2011 Jul 1;17(13):4550-7. Epub 2011 Apr 15.
Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy.
Rosenberg SA, Yang JC, Sherry RM, Kammula US, Hughes MS, Phan GQ, Citrin DE, Restifo NP, Robbins PF, Wunderlich JR, Morton KE, Laurencot CM, Steinberg SM, White DE, Dudley ME.
Source
Surgery Branch, National Cancer Institute, NIH, Bethesda, Maryland, USA. sar@nih.gov
Abstract
PURPOSE:
Most treatments for patients with metastatic melanoma have a low rate of complete regression and thus overall survival in these patients is poor. We investigated the ability of adoptive cell transfer utilizing autologous tumor-infiltrating lymphocytes (TIL) to mediate durable complete regressions in heavily pretreated patients with metastatic melanoma.
EXPERIMENTAL DESIGN:
Ninety-three patients with measurable metastatic melanoma were treated with the adoptive transfer of autologous TILs administered in conjunction with interleukin-2 following a lymphodepleting preparative regimen on three sequential clinical trials. Ninety-five percent of these patients had progressive disease following a prior systemic treatment. Median potential follow-up was 62 months.
RESULTS:
Objective response rates by Response Evaluation Criteria in Solid Tumors (RECIST) in the 3 trials using lymphodepleting preparative regimens (chemotherapy alone or with 2 or 12 Gy irradiation) were 49%, 52%, and 72%, respectively. Twenty of the 93 patients (22%) achieved a complete tumor regression, and 19 have ongoing complete regressions beyond 3 years. The actuarial 3- and 5-year survival rates for the entire group were 36% and 29%, respectively, but for the 20 complete responders were 100% and 93%. The likelihood of achieving a complete response was similar regardless of prior therapy. Factors associated with objective response included longer telomeres of the infused cells, the number of CD8(+)CD27(+) cells infused, and the persistence of the infused cells in the circulation at 1 month (all P(2) < 0.001).
CONCLUSIONS:
Cell transfer therapy with autologous TILs can mediate durable complete responses in patients with metastatic melanoma and has similar efficacy irrespective of prior treatment. Clin Cancer Res; 17(13); 4550-7. ©2011 AACR.
Contego core technology - Patient selection.
The first step in the Contego therapy will be the collection of the "TIL" (tumor infiltrating lymphocytes) if you don't know what these are you are a dummy investor - pardon me.
The second step in the process is growing the cancer-killing TIL in the laboratory. I noticed this research abstract published this year. It seems mundane, but looks like an physician investor would want to see these data. This article describes the features of the patients that will most likely be able to grow TIL. As most savvy investors in cancer now know, clinical trial are dependent on patients selection -this is the basis of personalized therapy. There is no reason for a person to enter a trial if there is not a reasonable expectation that they will benefit, the days of placebo control are over.
Back to the point. The second step is growing the cancer-killing TIL in the laboratory. By testing 226 patients in this step, the group at MD Anderson observed that there is near certainty of success at this step if the patient is under 30 years old. They also noticed that if the patient has received previous therapy, they are less likely to be successful at this step of the process. Women seem to to slightly better than men, which is interesting...
Also on a side note- two of the authors of this report are on the scientific advisory board. Hwu and Radvanyi. If there was any question as to the contribution of the board members, I am certainly satisfied. The selection criteria for a clinical trial is a critical aspect of the design. And the criteria described in this report are based on actual testing and not simply their opinions.
It would certainly be helpful for me if Genesis would highlight reports like this on their website. But I can only speculate that this kind of detail is reserved for high-level investor meetings with full presentation.
Peer-Reviewed scientific report
Clin Cancer Res. 2011 Jul 15;17(14):4882-91. Epub 2011 Jun 1.
Impact of clinical and pathologic features on tumor-infiltrating lymphocyte expansion from surgically excised melanoma metastases for adoptive T-cell therapy.
Joseph RW, Peddareddigari VR, Liu P, Miller PW, Overwijk WW, Bekele NB, Ross MI, Lee JE, Gershenwald JE, Lucci A, Prieto VG, McMannis JD, Papadopoulos N, Kim K, Homsi J, Bedikian A, Hwu WJ, Hwu P, Radvanyi LG.
Source
Department of Melanoma Medical Oncology, The University of Texas, M.D. Anderson Cancer Center, Houston, Texas 77030, USA.
Abstract
PURPOSE:
Clinical trials on adoptive T-cell therapy (ACT) using expanded tumor-infiltrating lymphocytes (TIL) have shown response rates of over 50% in refractory melanoma. However, little is known how clinical and pathologic features impact TIL outgrowth isolated from metastatic melanoma tumors.
EXPERIMENTAL DESIGN:
We analyzed the impact of clinical and pathologic features on initial TIL outgrowth in 226 consecutive patients undergoing tumor resection. Successful initial TIL outgrowth was defined as =40 million viable lymphocytes harvested from all tumor fragments in a 5-week culture. To normalize for the different size of resected tumors and thus available tumor fragments, we divided the number of expanded TIL by the starting number of tumor fragments (TIL/fragment).
RESULTS:
Overall, initial TIL outgrowth was successful in 62% of patients, with patients =30 years of age (94%; P = 0.01) and female patients (71% vs. 57% for males; P = 0.04) having the highest rate of success. Systemic therapy 30 days before tumor harvest negatively impacted initial TIL outgrowth compared to patients who never received systemic therapy (47% vs. 71%, P = 0.02). Biochemotherapy within 0 to 60 days of tumor harvest negatively impacted the initial TIL outgrowth with a success rate of only 16% (P < 0.0001).
CONCLUSION:
Parameters such as age, sex, and the type and timing of prior systemic therapy significantly affect the success rate of the initial TIL outgrowth from tumor fragments for ACT; these parameters may be helpful in selecting patients for melanoma ACT.
Scientific advancement in the core Contego Technology
I'll explain why this caught article my eye today. If you have read my previous posts you will notice that I have researched the Contego therapy. The first step for a patient to receive the "Contego therapy" at the NIH or MD Anderson Cancer Center is to have a tumor nodual surgically removed and sent to the processing lab. This is not unlike Dendreon's Provenge therapy, in the fact that it is personalized cellular therapy. However, Provenge requires the collection of blood, with the use of a special machine, like that used to collect platelets. Both of these processes are a barrier for the scale-up of the therapy.
This article published last week opens the door for Contego to be much more widely available. Whereas surgery has been the first step, now the first step can be a simple needle biopsy- no surgery means that the cost of producing the Contego therapy for each patent will drop dramatically, and that patients might be able to purchase this therapy and start treatment as their regional or even local hospitals.
This is exciting, because as you know if you've experienced cancer yourself or with a loved one, are a physician or in the medical field: the core-needle biopsy is typically used as a diagnostic procedure. Besides the 50-100 fold cost savings compared to surgery, the discomfort is much less and the stress compared to surgery is important for the patient.
Peer-reviewed scientific literature
Cancer Immunol Immunother. 2011 Dec 23.
Adoptive T-cell therapy for malignant melanoma patients with TILs obtained by ultrasound-guided needle biopsy.
Ullenhag GJ, Sadeghi AM, Carlsson B, Ahlström H, Mosavi F, Wagenius G, Tötterman TH.
Source
Department of Oncology, Radiology and Radiation Science, Uppsala University, 751 85, Uppsala, Sweden, Gustav.Ullenhag@onkologi.uu.se.
Abstract
Adoptive cell therapy with tumor-infiltrating lymphocytes (TIL) can mediate objective responses in up to 50% of malignant melanoma patients with a good performance status refractory to standard treatments. Current protocols for generation of TILs rely on open surgery for access to tumor tissue. We obtained tumor material by ultrasound-guided core needle biopsy or surgery from melanoma patients with progressive disease and were able to isolate >5 × 10(6) TILs from 23 of 24 patients who were subsequently treated with these cells. One-third of the individual TIL-positive cultures displayed interferon gamma activity after stimulation with relevant melanoma cell lines. When expanded TILs were used for treatment in combination with daily low dose s.c. IL-2 after prior lymphodepleting chemotherapy, we observed objective clinical responses in one patient treated with TILs obtained from surgery and 4 patients treated with TILs from core biopsies. The results of this study demonstrate for the first time the potential of core biopsies for generation of relevant numbers of TILs that can mediate objective responses in patients with metastatic malignant melanoma. Ultrasound-guided core needle biopsy is a robust, safe and inexpensive approach to obtain tumor tissue for TIL generation, and is especially valuable in instances where surgery is contraindicated.
Lets Separate these subjects
Regarding Til: forget about if it's effective. That doesn't matter. The question is, how many other companies are out there already providing Til, and ahead of the curve on GNBP?
There are no other companies offering Tils. And having a product with successful Phase 2 clinical from independent institutions does matter. This is not a preclinical development product. Although you seem to "know" the ceo lets agree to separate discussions of the governance from the product development if any. So as information is released people can discuss it on this board in the absence of heated personal opinions about executives.
Don't you think their CD-55 patent is the "Contego" brand name they concocted? I do.
CD55 antibody and Contego are two completly different products. Seems you don't know what you are talking about?
That's the issue with their CD-55 antibody treatment. Turns out CD-55 is widely manufactured and offers no competitive advantage for GNBP. I can buy CD-55 over the internet from many labs. GNBP doesn't even have the ability to make CD-55.
Most companies outsource the production of monoclonal antibody humanization, production and manufacture. But this has nothing to do with Contego as I mentioned before.
We also know a lot about Tony Cataldo, and this is a repeat of what he always does; find a distressed company with worthless patent, and re-market it to make it look like a new, exciting breakthrough product. Then it turns out the patent is either expiring, or is is the same thing available by many competitors, or is already obsolete because others have already marketed something better.
Til are a valuable therapeutic option. Just ask any melanoma patient, or reader of the New England Journal of Medicine, Science Magazine, or Journal of Clinical Oncology. When dendreon generated a cell therapy product for prostate cancer, they had to brand it with a name "Provenge". So the name is irrelevant. Though you should know what they are naming -as indicated above-
Unlike the other posts on this board that seem unhealthily obsessed with the ceo of this outfit, I've been researching the product Contego.
First there is no data from this company on Contego. Rather it seems that the new board members are the worlds experts on the therapy. When I looked at each of their Bios I see that they are working in the BEST cancer hospitals and have the most prestigious pedigrees.
I am shocked that the Genesis website does not have any information on the origin of Contego! The therapy was invented at the National Cancer Institute in Bethesda Maryland and is still the best therapy for Metastatic Melanoma available. It is actually call Til Therapy in the cancer world. that stands for Tumor Infiltrating Lymphocytes. It is very similar to Provenge from Dendreon.
Once I knew that Contego was just rebranding of Til, it was easy to find out about this product. These are clinical trials being run using Til/Contego currently, they are all
NCI/NIH
http://clinicaltrials.gov/ct2/results?term=til+melanoma+nih&recr=Open&no_unk=Y
MD Anderson
http://clinicaltrials.gov/ct2/results?term=til+melanoma+MD+anderson&recr=Open&no_unk=Y&rslt=&type=&cond=&intr=&outc=&lead=&spons=&id=&state1=&cntry1=&state2=&cntry2=&state3=&cntry3=&locn=&gndr=&rcv_s=&rcv_e=&lup_s=&lup_e=
What is interesting is that I was reading bulletin boards where melanoma patients discuss what therapy the are on, and they have a good impression of Til. What is strange is that there have already been Phase II clinical trials that have shown that this therapy works better the standard treatment which is high dose IL-2 (HD IL-2). So the question I have is " what does this company contribute to the equation?"
CASSIAN YEE, MD - MEMBER, SCIENTIFIC ADVISORY BOARD
Fred Hutchinson Cancer Research Center
http://www.fhcrc.org/research/profiles/yee.html
JAMES MULÉ, MD - MEMBER, SCIENTIFIC ADVISORY BOARD
Moffitt Cancer Center
http://www.moffitt.org/site.aspx?spid=AC68A6DF35AC4E9DB8150E413302996F
JEFFREY WEBER, MD - MEMBER, SCIENTIFIC ADVISORY BOARD
Moffitt Cancer Center
http://www.moffitt.org/site.aspx?spid=F0B14BB585AB4205A5798A3E718234A0
PATRICK HWU, MD - MEMBER, SCIENTIFIC ADVISORY BOARD
MD Anderson Cancer Center
http://faculty.mdanderson.org/patrick_hwu
DAVID DIGIUSTO, MD - MEMBER, SCIENTIFIC ADVISORY BOARD
City of Hope
http://www.cityofhope.org/directory/people/digiusto-david/Pages/default.aspx
DANIEL POWELL, MD - MEMBER, SCIENTIFIC ADVISORY BOARD
University of Pennsylvania School of Medicine
http://www.med.upenn.edu/apps/faculty/index.php/g5455356/p8186734