throbber
Chinese Journal of Cancer
`
`Immune checkpoints in cancer clinical trials
`
`Elad Sharon‘, Howard Streicher‘, Priscila Goncalves2 and Helen X. Chen1
`
`Abstract
`
`Immunology-based therapy is rapidly developing into an effective treatment option for a surprising
`
`range of cancers. We have learned over the last decade that powerful immunologic effector cells may
`
`be blocked by inhibitory regulatory pathways controlled by specific molecules often called "immune
`
`checkpoints." These checkpoints serve to control or turn off the immune response when it is no longer
`
`needed to prevent tissue injury and autoimmunity. Cancer cells have learned or evolved to use these
`
`mechanisms to evade immune control and elimination. The development of a new therapeutic class
`
`of drugs that inhibit these inhibitory pathways has recently emerged as a potent strategy in oncology.
`
`Three sets of agents have emerged in clinical trials exploiting this strategy. These agents are antibody-
`
`based therapies targeting cytotoxic T-lymphocyte antigen 4 (CTLA4), programmed cell death 1 (PD-1),
`
`and programmed cell death ligand 1 (PD-L1). These inhibitors of immune inhibition have demonstrated
`
`extensive activity as single agents and in combinations. Clinical responses have been seen in melanoma,
`
`renal cell carcinoma, non—small cell lung cancer, and several other tumor types. Despite the autoimmune
`
`or inflammatory immune-mediated adverse effects which have been seen, the responses and overall
`
`survival benefits exhibited thus far warrant further clinical development.
`
`Key words Immune checkpoints, PD-1, PD—L1, CTLA4, immunotherapy, immune modulators
`
`Immune checkpoints refer to regulatory pathways in the
`immunome that inhibit a portion of an active immune response
`against a specific target or set of targetsm. Immune checkpoints
`are therefore seen as a normal part of the immune system’s
`regulatory cascade and necessary to modulate and maintain immune
`homeostasis. These regulatory pathways are redundant, multi-
`faceted, and complex (Figure 1). Their presence and evolutionary
`development are thought to reflect the fact that a normal immune
`response can also be potentially deadly to a host if not properly
`regulated, as evidenced by autoimmune disease.
`As immunotherapy has gained a foothold in the anticancer
`armamentarium, there is a commonly held belief among
`oncologists that cancers thwart these regulatory mechanisms to
`evade immune detection and continue their growth and spread.
`However, the whole picture is more complex.
`In many chronic
`infections, immune checkpoints are exploited by parasitic and viral
`
`Authors' Affiliations: ‘Cancer Therapy Evaluation Program, Division of
`Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda,
`MD 20892, USA; 2Medical Oncology Service, Center for Cancer
`Research, National Cancer Institute, Bethesda, MD 20892, USA.
`Corresponding Author: Elad Sharon, 9609 Medical Center Drive,
`5W502, Bethesda, MD 20892, USA. Tel: +1-240-276-6565; Fax: +1-
`240-276-7894; Email: sharone@mail.nih.gov.
`dol: 10.5732/cjc.014.10122
`
`pathogens as evidenced by T-cell exhaustion in these conditions“.
`Cancer, however, arises from within the host. While some immune
`checkpoints are engaged to avoid immune-mediated eradication,
`others are merely a part of the normal immune response to stimuli.
`We are only beginning to understand these pathways.
`As a therapeutic class, drugs that inhibit these co-inhibitory
`signaling pathways, also known as immune checkpoint inhibitors,
`have emerged as a mainstay in melanoma therapy, with potential
`roles in the treatment of renal cell carcinoma (RCC), non—small cell
`lung cancer (NSCLC), urothelial cancer, head and neck cancer,
`ovarian cancer, and various lymphomasm”. In fact, ClinicalTrials.
`gov now lists dozens of trials with immune checkpoint inhibitors in a
`broad range of indications. As the targets of therapy are components
`of the immune system, there is currently no theoretical reason
`to exclude any potential histologies or tumor types from clinical
`evaluation. However, from a drug development perspective, this can
`be a challenge, as the resources are constantly constrained. It will
`be important to optimize the development of strategies to efficiently
`explore the therapeutic potentials.
`While a variety of agents could be deemed to interact with
`immune checkpoint inhibitors, the focus of this review are limited
`to the most advanced agents in clinical trials, those targeting the
`cytotoxic T-lymphocyte antigen 4 (CTLA4), programmed cell death 1
`
`434 www.cjcsysu.com
`
`CA CA Chinese Anti-Cancer Association
`Genome Ex. 1016
`
`Page 1 ofll
`
`Genome Ex. 1016
`Page 1 of 11
`
`

`

`Elad Sharon et al.
`
`Immune checkpoints in cancer clinical trials
`
`(PD-1), and programmed cell death ligand 1 (PD-L1) pathway. The
`early trials involving CTLA4- and PD-1lPD—L1—targeting agents have
`shown how the addition of just one blocking antibody can reveal
`a hidden immune response, with potentially massive therapeutic
`benefit for patients. Immune modulators targeting other mechanisms
`(Table 1). such as indoleamine 2,3-dioxigenase (IDO)“2'13],
`
`lymphocyte-activation gene 3 (LAG3)“‘“, T-cell membrane protein 3
`(TIM3)M, signal transducer and activator of transcription 3 (STAT3)“6],
`glucocorticoid-induced tumor necrosis factor receptor (TNFR) family-
`related protein (GITR)“7], and agents against the human inhibitory
`killer IgG-like receptor (anti-KIR) targeting natural killer (NK) cells
`have also entered the clinical trials arenanml.
`
`Lu mph node.-
`
`Peripheral ti55 Lie
`
`
`
`TL'R Mlli'
`II-
`
`PL]
`
`1
`
`I‘L'I-LI
`
`.! I1| i -F‘[}-'|
`
`L arl'l'J’IJ-L'I
`
`Jammie
`I
`
`NK cell.
`
`.Inli (Tin-1
`
`Figure 1. Regulatory pathways In Immune-oncology. The immune system has multiple levels of co-stimulatory (shown
`in green) and co-inhibitory (shown in red) pathways, helping to maintain immune homeostasis in the midst of responding
`to antigenic stimulation. Antibodies blocking cytotoxic T-lymphocyte antigen 4 (CTLA4), programmed cell death 1 (PD-1),
`and programmed cell death ligand 1 (PD-L1) have shown remarkable clinical activity and further development is underway.
`Agonist antibodies, which directly interact with co-stimulatory pathways such as 41 BB and CD40, are also in clinical
`development. Other means of affecting the immune responses are being explored, including direct actionon regulatory T (Treg)
`cells and natural killer (NK) cells. MHC, major histocompatibility complex; TCR, T-cell receptor.
`
`Table 1. Other immunotherapeutic agents in development
`
`Target
`
`Name
`
`lndication(s)
`
`Company
`
`Phase
`
`Clinical Trials.gov identifier (selected trials)
`
`Heme, hematologic tumors; ATL, acute T-cell leukemia; CTCL, cutaneous T-cell lymphoma; AML, acute myeloid leukemia.
`
`87.1
`B?H3
`LAGS
`
`CD137
`
`KIR
`
`CCR4
`CDZ7
`0x40
`
`CD40
`
`Galiximab
`MGA271
`IMP321
`EMS-986016
`BMS-663513
`PF—05082566
`IPH2101
`
`KW-0761
`CDX—1127
`MEDl-6469
`
`CP-870,893
`
`Lymphoma
`Solid tumors
`Solid tumors
`Solid tumors
`Solid tumors
`Lymphoma
`Myeloma, AML
`
`Biogen Idec
`Macrogenics
`lmmuntep
`Bristol-Myers Squibb
`Bristol-Myers Squibb
`Pfizer
`Innate Pharma (BMS)
`
`Kyowa Kirin
`ATL, CTCL
`Solid tumors & Heme CellDex Therapeutics
`Solid tumors
`Medlmmune/AZ
`
`Pancreatic
`
`Genentech
`
`Phase II
`Phase I
`Phase II"
`Phase I
`Phase II”
`Phase I
`Phase II
`
`Phase l/ll
`Phase I
`Phase I
`
`Phase I
`
`NCT00516217
`NCT01391143
`NCT00349934
`NCT01968109
`NCT00309023
`NCT01307267
`NCT01248455
`NCT01256073
`
`NCT00920790
`NCT01460134
`NCT02205333
`
`NCT01456585
`
`www.cjcsysu.com
`
`Chin J Cancer; 2014; Vol. 33 Issue 9 435
`Genome Ex. 1016
`
`Page 2 of 11
`
`Genome Ex. 1016
`Page 2 of 11
`
`

`

`Elad Sharon et al.
`
`Immune checkpoints in cancer clinical trials
`
`CTLA4 Inhibition and Human Cancers
`
`CTLA4 background
`
`CTLA4 is one of several co-inhibitory molecules that aid in
`modifying the T-cell response to antigen activation. The primary role
`of CTLA4 is to modify T-lymphocyte response to stimuli. CTLA4
`regulates the clonal burst size during priming and secondary
`expansion, which is thought to be proportional to the activation
`strength of the complex formed by the T-cell receptor—major
`histocompatibility complex (TCR—MHC complex), both in terms of
`binding affinity and co-stimulation by accessory signals. CTLA4,
`also known as cluster of differentiation (CD) 152, is an inhibitory
`molecule found on T cells, and its counterpart is CD28. CD28 is a co-
`stimulatory signal, but it is important to note that CTLA4 has higher
`avidity for its ligands than CD28, which suggests a dominance of
`inhibitory signals in immune activation. The ligands for both CD28
`and CTLA4 are known as B7 proteins, which are found on antigen-
`presenting cells (APCs). There are two types of B7 proteins: B7-1 (also
`known as CD80) and B7-2 (also known as CD86)[21'22].
`In normal circumstances, T lymphocytes are thought to express
`CTLA4 on their surface immediately upon response to antigenic
`stimulation of TCR. Because a blocking antibody would attenuate
`the inhibitory signal, an anti-CTLA4 antibody would appear to have a
`clear role in enhancing antitumor immunity, as the interaction of B7
`with CD28 would thereby be enhanced. Indeed, in preclinical models,
`this is exactly what was seenlz’]. However, B7 is rarely present in the
`tumor microenvironment, leading to a second hypothesis for how anti-
`CTLA4 antibodies may enhance immune-mediated tumor rejectionm'.
`CTLA4 is differentially expressed among separate T-Iymphocyte
`subsets. Indeed, higher levels of surface expression of CTLA4 are
`seen in regulatory T cells (Treg cells) as compared with effector
`T cells (Teff cells). Researchers have established that higher Teffl
`Treg ratios in both CD4—positive and CD8-positive tumor-infiltrating
`lymphocytes are associated with enhanced tumor eradication when
`CTLA4 blockade is employed in tumor modelslz‘m.
`Other mechanisms of action involving Treg cells have been
`hypothesized. For example, Treg cells are known to reduce B7
`expression on both human and animal APCSW. While B7 is thought
`to directly signal CTLA4, leading to its co-inhibitory effect, the reverse,
`or CTLA4 signaling to APCs via B7, also occurs. CTLA4 reverse
`signaling via B7 can lead to increased expression of IDO. IDO leads
`to a reduced level of local tryptophan stores in the microenvironment,
`which inhibits T-lymphocyte activation and proliferationm. Lastly,
`recently discovered alternatively spliced mRNA molecules encode
`soluble CTLA4 molecules lacking transmembrane domains. These
`soluble inhibitors could also mediate immune suppression, either by
`down-regulating B7 expression as noted above or by blocking the
`potential interaction of B7 and the co-stimulatory molecule CD28”.
`
`lpilimumab
`
`The first anti-CTLA4 agent in clinical development, ipilimumab,
`was approved by the United States Food and Drug Administration
`
`(FDA) and the European Medicines Agency for the treatment
`of metastatic melanoma following research showing improved
`survivalm]. lpilimumab is a fully humanized immunoglobulin (lg) G1
`kappa monoclonal antibody that antagonizes CTLA4 and prevents
`ligand binding”.
`A phase III combination study of ipilimumab at 3 mglkg with a
`glycoprotein 100 (gp100) peptide vaccine was conducted with 676
`patients with stage III or IV unresectable melanoma, randomized in
`a 3:1 :1 ratio. The results showed the median overall survival (OS) in
`the ipilimumab plus gp100 group was 10.0 months [95% confidence
`interval (CI), 8.5 to 11.5 months], as compared with 6.4 months
`(95% CI, 5.5 to 8.7 months) in the gp100 alone group [hazard ratio
`(HR) for death, 0.68; P < 0.001]. The median OS in the ipilimumab
`alone group was 10.1 months (95% CI, 8.0 to 13.8 months; HR for
`death with ipilimumab alone as compared with gp100 alone, 0.66;
`P = 0.003). Grade 3 or 4 immune-related adverse events occurred
`in 10% to 15% of patients treated with ipilimumab and in 3% of
`patients treated with gp100 alone. The investigators concluded that
`ipilimumab, with or without a gp100 peptide vaccine, as compared
`with gp100 alone, improved OS in patients with previously treated
`metastatic melanoma. Adverse events can be severe, long-lasting, or
`both, but most are reversible with appropriate treatment, particularly
`corticosteroids”.
`
`Another supportive study in metastatic melanoma involved 502
`patients in a 1:1 randomized trial of ipilimumab at a dose of 10 mglkg
`plus dacarbazine at a dose of 850 mg/m2 of body surface area versus
`dacarbazine and placebo. The OS was longer in the group receiving
`ipilimumab-dacarbazine therapy than in the dacarbazine-placebo
`arm (11.2 months vs. 9.1 months). Survival rates were also higher
`for the ipilimumab-dacarbazine arm at 1 year (47.3% vs. 36.3%),
`2 years (28.5% vs. 17.9%), and 3 years (20.8% vs. 12.2%), with
`an HR for death at 0.72 (P < 0.001). Grade 3 or 4 adverse events
`were recorded in 56.3% of patients in the experimental arm and only
`27.5% in the control arm“. These findings further support the use of
`ipilimumab in metastatic melanoma.
`The preliminary results of the first adjuvant trial were reported
`at the 2014 Annual Meeting of the American Society of Clinical
`Oncology (ASCO). The European Organization for Research and
`Treatment of Cancer (EORTC) conducted a trial with 951 patients
`with stage III melanoma following complete resection (EORTC 18071,
`NCT00636168). Patients were randomized to receive ipilimumab at
`a dose of 10 mglkg versus placebo every 3 weeks for 4 doses. The
`median relapse-free survival was 26.1 months for the ipilimumab arm
`and 17.1 months for the placebo arm, with an HR of 0.75 (95% CI,
`0.64 to 0.90; P = 0.001). The adverse event profile was thought to
`be generally consistent with that seen in advanced melanoma, but a
`higher incidence of endocrinopathies were reported“.
`While the results were largely seen as positive, there are still
`unresolved issues that may limit the ability of regulatory agencies
`to approve ipilimumab in the adjuvant setting. In particular, patients
`with stage III melanoma have the option of high-dose interferon,
`which is the standard of care in the United States“. In addition, as
`noted above, the dose chosen in EORTC 18071 trial was 10 mglkg
`of ipilimumab, higher than the dose used in the metastatic setting.
`
`436 Chin J Cancer; 2014; Vol. 33 Issue 9
`
`Chinese Journal of Cancer
`
`Genome Ex. 1016
`
`Page 3 of 11
`
`Genome Ex. 1016
`Page 3 of 11
`
`

`

`Elad Sharon et al.
`
`Immune checkpoints in cancer clinical trials
`
`Another large adjuvant trial, E1609 (NCT01274338), addresses the
`questions of the optimal dose of ipilumumab and its comparative
`effectiveness to high-dose interferon.
`In this trial, conducted by the
`Eastern Cooperative Oncology Group (ECOG) under the sponsorship
`of the United States National Cancer Institute (NCI), 1,500 patients
`with stage III or IV melanoma that has been fully resected are
`randomized to three arms: high-dose interferon, ipilimumab at 3 mg/
`kg, and ipilimumab at 10 mglkgm]. The trial is close to completing
`accrual and the outcome data are eagerly awaited.
`Other indications with ipilimumab are still under clinical
`development. On ClinicalTrials.Gov, a search with the term
`“ipilimumab" returns 191 trials, over 100 of which remain open. Much
`of the attention in ipilimumab clinical development has moved to
`combination therapy, particularly with the anti-PD-1 agent nivolumab.
`The development of ipilumumab brought up interesting ques-
`tions about the best endpoints of efficacy assessment for this class
`of agents. In the phase III melanoma trials with ipilimumab described
`above, improvements in OS seen in the metastatic setting were not
`accompanied by significant radiographic responses to therapy. This
`has complicated efficient drug development, as response rate is
`less likely to serve as a readout of efficacy, at least when ipilimumab
`is used as a monotherapy. Monotherapy is still being explored in
`various settings, however.
`In one example, in a single institution
`case series, 5 patients with recurrent glioblastoma multiforme (GBM)
`who were treated with ipilimumab were followed. Patients had time
`to progression ranging from 1 to 6 months, and 1 patient remains
`recurrence-free 19 months after therapy, suggesting penetration of
`this drug across the blood-brain barrierm].
`Currently, there are no biomarkers to predict which patients or
`tumor types are more likely to respond to anti-CTLA4 therapies.
`Some analysis suggested that a high mutational load may be
`associated with a clinical benefit from ipilimumab.
`In particular,
`one group of researchers has focused on identifying a “neoantigen
`signature” that correlates with benefit. Findings are preliminary,
`but this suggests that tumor genetics might explain the divergent
`outcomes among patients treated with ipilimumablas'.
`
`Tremelimumab
`
`Tremelimumab (formerly CP-675,206) is a human lgG2
`monoclonal antibody specific for CTLA4. In a large, single-arm phase
`II trial, 241 response-evaluable patients with advanced refractory or
`relapsed melanoma were treated at a dose of 15 mglkg intravenously
`every 3 months. Objective responses were observed in 6.6% of those
`patients, which led to a phase III trial in a similar patient population“.
`In the phase III trial in advanced melanoma, 655 patients were
`enrolled and randomly assigned to treatment with tremelimumab or
`chemotherapy. Median OS by intent to treat was 12.6 months (95%
`CI, 10.8 to 14.3 months) for tremelimumab and 10.7 months (95%
`Cl, 9.36 to 11.96 months) for chemotherapy (HR, 0.88; P = 0.127).
`Of note, while objective response rates were similar in the two arms
`(10.7% in the tremelimumab arm and 9.8% in the chemotherapy
`arm), duration of response was significantly longer among patients
`treated with tremelimumab (35.8 months vs. 13.7 months, P =
`
`www.cjcsysu.com
`
`0.0011)“ Regardless, the study failed to demonstrate a statistically
`significant survival advantage of treatment with the investigational
`drug over standard-of-care chemotherapy.
`While the results were disappointing, there was a possibility that
`the dosing schedule of tremelimumab (once every 3 months) was
`insufficient to achieve results similar to that of ipilimumab. As a result,
`ongoing efforts with tremelimumab as a monotherapy have used more
`frequent dosing. The preferred dose and schedule for tremelimumab
`for ongoing trials has generally been 10 mglkg intravenously, every
`4 weeks for 6 months. A phase II, single-ann study of tremelimumab
`as a second-line treatment was conducted with 29 patients with
`malignant mesothelioma. Results showed a partial response in 4
`patients (13.8%) and stable disease in 11 patients (37.9%), with OS
`of 11.5 months“. As a result, a phase III, double-blinded, randomized
`trial is currently undenlvay using tremelimumab in mesotheliomam].
`Tremelimumab is also being tested in various combinations in a
`variety of other tumor types.
`
`PD-1lPD-L1 Inhibition and Human
`Cancers
`
`PD-l/PD-Ll background
`
`Compared to the limited development with CTLA4, there is
`an immense level of interest and investment by pharmaceutical
`companies in the PD-1/PD-L1 pathway (Table 2). Seven major
`companies have entered the fray, with multiple phase III registration
`trials already underway in a variety of malignancies, most notably
`melanoma, RCC, and NSCLC (Table 3). This enthusiasm is justified,
`as clinical trial results thus far confirm that targeting the PD-1/PD-
`L1 pathway represents the most exciting opportunity for advancing
`cancer immunotherapy to date.
`Whereas CTLA4 is involved in central tolerance and control, the
`PD-1 receptor is critical in peripheral tolerance. PD-1 is expressed on
`T lymphocytes during thymic development. Similar to CTLA4, PD-1
`becomes expressed on CD4— and CD8—positive T lymphocytes during
`antigenic stimulation, serving as a co-inhibitory signal. In addition,
`PD-1 is expressed on numerous other immune cells, including
`natural killer T cells, B cells, monocytes, and certain dendritic cell
`subsets“°"‘”. PD-1 has two main ligands, namely PD-L1 (also known
`as B7-H1) and PD-L2 (also known as B7-DC). While PD-L2 has a
`much higher affinity for PD-1,
`it is expressed chiefly on activated
`dendritic cells, macrophages, certain B-cell subsets, and other
`immune cells. PD-L1 is more widely expressed on hematologic and
`non-hematologic tissuesm.
`As a co-inhibitory signal, PD-1 engagement results in reduced
`cytokine production, cytolytic activity, and lymphocyte proliferationl‘”.
`PD-1 is up—regulated in T lymphocytes following viral infections and
`down-regulated following viral clearance. In chronic viral infections,
`however, CD8-positive T lymphocytes express PD-1 constitutively,
`likely through gene demethylation, contributing to what has been
`termed a T-cell exhaustion phenotype. In human immunodeficiency
`virus (HIV) infection, for example, high levels of PD-1 expression
`allow for persistent interaction between PD-L1 expressed by APCs
`
`Chin J Cancer; 2014; Vol. 33 Issue 9 437
`Genome Ex. 1016
`
`Page 4 of 11
`
`Genome Ex. 1016
`Page 4 of 11
`
`

`

`Elad Sharon et al.
`
`Immune checkpoints in cancer clinical trials
`
`Table 2. Agents targeting PD-1/PD-L1 in clinical development
`
`Company
`
`Agent targeting PD-1
`
`Agent targeting PD-L1
`
`CT-01 1
`
`Pidilizumab (humanized lgG1 mAb)
`
`Medlmmune/AZ
`
`AMP-514
`
`MEDI4736 (fully human mAb)
`
`EMD Serono
`
`N/A
`
`MSBDO107180
`
`PD-1, programmed death 1 receptor; PD-L1, programmed cell death ligand 1;
`available.
`
`lgG4, immunoglobulin G4; mAb, monoclonal antibody; N/A, not
`
`NCT02008227
`
`Table 3. Selected ongoing PD-1/PD-L1 trials in melanoma, renal cell cancer (RCC), and non—small cell lung cancer
`(NSCLC)
`
`Sponsor
`
`Setting
`
`Comparison
`
`Primary
`endpoint
`
`Clinical trials.
`Sample Primary
`size
`completion date gov identifier
`
`Bristol-Myers Squibb NSCLC
`
`3rd-line squamous cell
`
`||
`
`Nivolumab vs. docetaxel
`
`OS
`
`264
`
`Jan 2016
`
`NCT01642004
`
`Merck
`
`Melanoma
`
`ist-/2nd-Iine (ipi-nai've)
`
`III
`
`Bristol-Myers Squibb Melanoma 2nd-Iine post-ipi
`
`Bristol-Myers Squibb Melanoma 1st-/2nd-line
`
`Genentech
`
`NSCLC
`
`2nd-line
`
`Genentech
`
`NSCLC
`
`2nd-Iine
`
`III
`
`|||
`
`II
`
`III
`
`Pembrolizumab vs.
`ipilimumab
`
`Nivolumab vs.
`chemotherapy
`
`Nivolumab vs.
`chemotherapy
`
`OS & PFS 645
`
`Feb 2015
`
`NCT01866319
`
`OS
`
`405
`
`May 2015
`
`NCT01721746
`
`OS
`
`410
`
`Sept 2015
`
`NCT01721772
`
`MPDL3280A vs. docetaxel OS
`
`287
`
`Mar 2016
`
`NCT01903993
`
`MPDL3280A vs. docetaxel 03
`
`850
`
`Jun 2017
`
`ORR, overall response rate; OS, overall survival; PFS, progression-free survival; ipi, ipilumumab. Other footnotes as in Table 2.
`
`and subsequent T-cell dysfunction. T-lymphocyte activation is
`essentially blocked in this setting. In an experiment using B cells from
`HIV-infected individuals, researchers were able to show increased
`responses to HIV antigens in the presence of PD-1 blocking
`antibodies in vitro‘m. This suggests that an active immune response
`can be induced if effective blockade of the PD-1IPD-L1 pathway
`can be implemented. A clinical trial sponsored by the US National
`Institutes of Health is currently planned to investigate that possibility
`
`using the anti-PD-L1 antibody, BMS—936559 (NCT02028403).
`Similarly, tumors use the same pathway, meant to induce
`peripheral immune tolerance, to evade T-lymphocyte-mediated
`immune eradication. Tumors evading an active immune response
`are thought to express PD-L1 following T lymphocyte infiltration
`and expression of interferon-gamma. Expression of PD-L1 has
`been associated with a poor prognosis in a wide variety of human
`tumorsm'm”. Interestingly, in one analysis of glioma cases, a better
`
`438 Chin J Cancer; 2014; Vol. 33 Issue 9
`
`Chinese Journal of Cancer
`
`Genome Ex. 1016
`
`Page 5 ofll
`
`Genome Ex. 1016
`Page 5 of 11
`
`

`

`Elad Sharon et al.
`
`Immune checkpoints in cancer clinical trials
`
`prognosis was seen in patients whose tumor-adjacent brain tissue
`expressed PD-L1 and whose tumors did not express PD-L1. This
`suggests a general role for PD-L1 in protecting normal tissue from
`immune attack, though it would be important to see these findings
`replicated elsewhere and in other tumor typeslsz].
`In general, the results of PD-1/PD-L1 blockade have been
`encouraging. Higher response rates and durable responses were
`seen with PD-1/PD-L1 blocking antibodies than with CTLA4 blockade
`in melanoma, RCC, and unexpectedly, NSCLC. In addition, although
`similar immune-mediated adverse events were seen with both CTLA4
`
`and PD-1/PD-L1 agents, the severity of adverse events has generally
`been lower, with the caveat that fatal pneumonitis was seen in 1% of
`the patients on the nivolumab phase I trial described below“.
`
`PD-l—targeting agents
`
`Nivolumab
`
`Nivolumab (BMS-936558) is a fully human IgG4 monoclonal
`antibody targeting PD-1. A phase II” study was reported in 2012
`in the New England Journal of Medicine in parallel with a plenary
`session at the 2012 ASCO Annual Meeting“. Patients on the trial had
`advanced melanoma, NSCLC, castration-resistant prostate cancer,
`RCC, or colorectal cancer (CRC). Patients received nivolumab at
`doses of 0.1 to 10.0 mglkg of body weight every 2 weeks for up to 12
`cycles until disease progression or a complete response occurred.
`Of the 296 patients enrolled, only 14% experienced grade 3 or 4
`drug-related adverse events, but there were 3 deaths from pulmonary
`toxicity. No maximum tolerated dose (MTD) was defined. Cumulative
`response rates (all doses) were 18.4% (14/76) among patients with
`NSCLC. 27.6% (26/94) among patients with melanoma, and 27.3%
`(9/33) among patients with RCCW. A subsequent analysis of the
`104 patients in that trial with metastatic melanoma was performed.
`The investigators reported that the median OS in nivolumab-treated
`metastatic melanoma patients was 16.8 months, and 1- and 2-year
`survival rates were 62% and 43%, respectively. Among the 33
`patients (30.8%) with objective tumor regressions, the Kaplan-Meier
`estimated median response duration was 2 years. Of note, responses
`were not observed in prostate cancer and colon cancer patients on
`the trialls‘".
`
`While preliminary results from the phase II” trial suggested
`that PD-L1 could be an appropriate biomarker for patient selection,
`subsequent analyses have shown numerous PD-L1—negative
`patients had responded to treatment with nivolumab, although lower
`rates are seen. Given the variability and limitation of the available
`PD-L1 assays, a true absence of PD-L1 expression in those patients
`cannot be confirmed. At this time it remains to be determined whether
`
`PD-L1 is a predictive marker of response for PD-1 pathway inhibitors.
`At the 2014 ASCO Annual Meeting, preliminary results of other
`nivolumab clinical trials were reported. In a small dose-escalation trial
`in Japan, 3 of 13 patients with platinum-resistant ovarian cancer had
`objective responses to nivolumab. The initial 10 patients were treated
`at a dose of 1 mglkg, and 2 responders were from that cohort. The
`subsequent cohort that received 3 mglkg had only 3 patients, but 1
`was a responder. These results, though preliminary, were promisingm.
`
`www.cjcsysu.com
`
`In addition, investigators reported early results using nivolumab at
`a dose of 3 mglkg every 2 weeks in patients with treatment-naive
`advanced NSCLC. The initial results on 20 patients revealed an
`objective response rate of 30%. Two patients had a greater than
`80% target lesion reduction at 18 weeks. Of the 15 evaluable tumor
`samples, 9 were PD-L1—positive (defined as greater than 5% PD-
`L1 expression using a Dako kit), and the response rate was 67% in
`PD-L1—positive patients; whereas no responses were observed in
`the 6 PD-L1—negative patientsl55‘. Combinations of nivolumab with
`conventional chemotherapy or epidermal growth factor receptor
`(EGFR) inhibitors were also reported. but results are difficult to
`interpret in the absence of randomized comparisonslfi'sn.
`
`Pembrolizumab
`
`Pembrolizumab (MK-3475, formerly lembrolizumab) is a
`humanized IgG4 monoclonal antibody targeting PD-1. Pembro-
`lizumab has been very successful in treating melanoma and NSCLC,
`similar to nivolumab. Significant differences cannot be assessed
`in the absence of a randomized trial comparing the two agents.
`However, binding affinities of the agents are different. Nivolumab is a
`fully human IgG4, and pembrolizumab is humanized. In phase I trials,
`neither agent has been found to have a maximally tolerated dose.
`That said, more time and energy has been spent on searching for an
`appropriate dose for pembrolizumab.
`In the first major publication involving pembrolizumab, Hamid et
`al.” reported that patients with advanced melanoma were analyzed
`after being treated with three separate dosing strategies: 10 mg/
`kg of body weight every 2 or 3 weeks or 2 mglkg every 3 weeks.
`Ultimately, 135 patients with advanced melanoma were treated.
`Adverse events were similar to those found in patients treated with
`nivolumab, including fatigue, rash, pruritus, and diarrhea. Response
`rates across all dose levels were 38% (95% Cl, 25% to 44%).
`Investigators found no difference among those with and without
`prior ipilimumab therapy. Responses were durable, and the median
`progression-free survival (PFS) among the 135 patients was longer
`than 7 months“.
`
`A subsequent prospective, randomized analysis was performed
`using both the 2 mglkg and the 10 mglkg doses, given every 3 weeks
`to patients with ipilimumab-refractory advanced melanoma. There
`were 89 patients in the 2 mglkg cohort and 84 patients in the 10
`mglkg cohort. The response rate was 26% at both doses. Safety
`profiles were similar and there were no deaths reported”. Other
`attempts to analyze patients with melanoma have been reported,
`including an analysis of 411 melanoma patients treated across
`multiple dose levels and multiple trials, which was reported at the
`2014 ASCO Annual Meeting. Median OS data was not available, but
`1-year OS rate over all dose cohorts was 71%. Response rates were
`encouraging as well, ranging from 26% to 57%, varying based on
`ipilimumab prior treatment, dose, and schedule”.
`Investigators also reported preliminary results of a phase I trial
`of previously treated patients with locally advanced or metastatic
`NSCLC. Enrolled patients with PD-L1 detected in their tumors
`by a preliminary immunohistochemical assay were randomized
`to pembrolizumab at a dose of 10 mglkg every 2 weeks or every
`
`Chin J Cancer; 2014; Vol. 33 Issue 9 439
`Genome Ex. 1016
`
`Page 6 ofll
`
`Genome Ex. 1016
`Page 6 of 11
`
`

`

`Elad Sharon et al.
`
`Immune checkpoints in cancer clinical trials
`
`3 weeks. In addition, some patients with tumors without PD-L1
`expression who had received at least two prior lines of therapy were
`treated at a dose of 10 mglkg every 2 weeks. Ultimately, 102 patients
`comprised the every-2-week cohort (including 43 whose tumors did
`not express PD-L1), and 119 patients comprised the every-3-week
`cohort. Investigators reported that 48% of patients experienced drug-
`related adverse events, with 6% experiencing grade 3/4 adverse
`events. As in the nivolumab trial reported above, pneumonitis was
`a concern, with 3 cases of drug-related grade 3/4 pneumonitis. The
`Response Evaluation Criteria in Solid Tumors (RECIST) response
`rate in all patients was 21%. The response rate was slightly higher
`(24%) for patients with PD-L1—positive tumors (more specifically.
`31% on the every-2-week cohort and 22% on the every-3-week
`cohort). RECIST response rates were 8% for patients without PD-L1
`expression“.
`
`Pidilizumab
`
`Pidilizumab is a humanized lgG1 antibody targeting PD-1. The
`agent was initially evaluated in a phase I trial targeting hematologic
`malignancies. Presently, there are a number of clinical trials
`undenivay in both hematologic and solid tumors”.
`The results of two pidilizumab clinical trials were recently
`published in peer-reviewed journals. In a single-center, single-arm,
`phase II trial, 32 patients with relapsed follicular lymphoma received
`pidilizumab at a dose of 3 mglkg every 4 weeks for 4 infusions with
`up to 8 additional infusions administered. In addition, rituximab was
`given at a dose of 375 mg/m2 of body surface area every week for
`4 weeks. Investigators reported that 19 of 29 evaluable patients
`achieved an objective response, with complete responses in 15
`patients (51.7%)“.
`An additional phase II trial involved patients with diffuse large
`B-cell lymphoma (DLBCL) following autologous hematologic stem cell
`transplantation (AHSCT). Sixty-six patients were treated with 3 doses
`of pidilizumab in the first 1 to 3 months after AHSCT. The PFS rate
`was 72% at 6 months after AHSCT (90% CI, 60% to 82%), meeting
`the primary endpoint. Thirty-five patients had measurable disease
`following AHSCT, and the response rate in those patients was 51 %m.
`Investigators also presented results on the use of pidilizumab
`in metastatic melanoma at the 2014 ASCO Annual Meeting. In this
`trial, 103 patients were randomized in a 1:1 ratio to receive either
`1.5 mglkg or 6 mglkg every 2 weeks for 27 doses. Response rates
`were lower than those observed

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