Is There a Role for Radiation Therapy and Immunotherapy?

Total Page:16

File Type:pdf, Size:1020Kb

Is There a Role for Radiation Therapy and Immunotherapy? IS THERE A ROLE FOR RADIATION THERAPY AND IMMUNOTHERAPY? S. Lewis Cooper, M.D. Assistant Professor, Radiation Oncology Hollings Cancer Center Medical University of South Carolina (MUSC) DISCLOSURE Abney Clinical Scholar OBJECTIVES • Review mechanisms of immune escape by cancer cells • Review radiation effects on the immune system • Review current strategies to combine RT and immunotherapy • IL-2 • Dendritic cell production • Tumor antigen vaccine • CTLA-4 antibody/PD-1antibody • TLR agonist • TGF-β antibody CANCER IMMUNOEDITING CD8+ NKT CD8+ T cell cell T cell • Three phases: NK CD4+ T cell • Elimination – Innate and adaptive immune systems detect INF-γ IL-12 and destroy developing tumor before it is clinically TNF TRAIL apparent. Mφ Perforin γδ T DC CD8+ IL-12 cell T cell CD4+ INF-γ T cell • Equilibrium – The immune system maintains residual tumor cells in a functional state of dormancy. Protection Mφ CD8+ NK T cell TGF-β PD-L1 IL-6, IL-10 • Escape – Tumor cells that aquire the ability to escape Galactin-1 IDO Antigen Loss immune recognition and destruction emerge as growing MHC Loss tumors. CD8+ T cell CTLA-4 CTLA-4 PD-1 PD-1 CD8+ MDSC Treg Science . 2011;331:1565-70 T cell MECHANISMS OF ESCAPE • Loss of tumor antigen expression 1. Tumor cells that do not express strong rejection antigens. 2. Loss of MHC class 1 proteins that present these antigens 3. Loss of antigen processing function • Immunosuppresive state in the tumor microenvironment 1. Production of immunosuppresive cytokines (VGEF, TGF-β, galectin, IDO) 2. Recruitment of immunosuppressive cells (Treg, MDSCs, TAMs) Science . 2011;331:1565-70 RADIATION EFFECTS ON THE IMMUNE SYSTEM • Upregulation of HLA, presentation of TAAs, FAS expression Vascular normalization Low • May restore immune effector recognition and immune- T cell infiltration mediated cell death Increase peptides • Skewing of cytokines to inflammatory repertoire; Increase ICAM-1 and FAS RT upregulation of co-stimulatory molecules Upregulate MHC-1 Dose • Upregulation of chemokines, adhesion molecules Chemokine release (VCAM-1, E-selectin, ICAM-1) Activation and expansion • Restoration of regulated APC trafficking of tumor specific CD4 and CD8 T cells • Upregulation of co-stimulatory molecules Increased dendritic cell High • Suppressive - Increase Tregs and activate TGF-β uptake and presentation of TAA (Calreticulin, HMGB1) PNAS. 1989;86:10104-7 J Immunol. 2003;170:6338-47 Cancer Res. 2004:64:7985-94 J Immunol. 2008;180:3132-9 Nat Med. 2007;13:1050-9 Front Oncol. 2012;2:90 J Clin Invest. 1994;93:892-9 EARLY EVIDENCE • 3-methylcholanthrene-induced fibrosarcoma (FSa) • TCD50 – Radiation dose to control 50% of tumors • Normal syngeneic C3Hf/Bu mice TCD50 • Mice with 600 rad whole body irradiation 7000 (WBI) 6000 • Mice permanently immunosuppressed with 5000 thymectomy and 900 rads WBI followed by 4000 syngeneic bone marrow (TxIR) reconstitution. 3000 Rads 2000 • Mice treated with Corynebacterium parvum 1000 0 Control 600 WBI TxIR C. parvum J Natl Cancer Inst . 1979;63:1229-35 RT DOSE AND FRACTIONATION RT DOSE & FX AND IMMUNE RESPONSE • WT or nude mice injected with B16 melanoma cells 20 Gy x 1 • Increasing immunogenicity of B16 cells did not influence RT mediated regression. • CD8 + 2C transgenic cells CFSE labeled and transferred into mice. 15 Gy x 3 15 Gy x 3 Blood . 2009;114:589-95 RT DOSE & FX AND IMMUNE RESPONSE • Chemotherapy and fractionated RT diminish the effect of RT ablation and + B16 melanoma 4T1 mammary CD8 priming. carcinoma • RT + Ad-LIGHT immunotherapy 15 Gy x 3 Paclitaxel reduces lung metastases 20 Gy x 1 Dacarbazine 12 Gy x 2 Blood . 2009;114:589-95 PRE-CLINICAL AND CLINICAL EVIDENCE IL-2 Mφ IL-2 and TIL CD8+ NK T cell TGF-β PD-L1 IL-6, IL-10 Galactin-1 IDO RT CD8+ T cell CTLA-4 CTLA-4 PD-1 PD-1 CD8+ MDSC Treg T cell IL-2, TIL, +RT • MC-38 adenocarcinoma liver metastases • RT effect thought to be to direct anti-tumor activity. • IL-2 alone activity with local RT. • Dose dependent effect of local RT • Treating ½ liver showed no anti-tumor activity with IL-2 and RT RT J Exp Med . 1990;171:249-63 PILOT STUDY OF RT AND IL2 • Metastatic cancer with at least two sites of n=28 measurable disease. Median Age (range), yrs 48.5 (26-66) Sex Surgery for obtaining tumor Male 21 (75%) for TIL preparation Female 7 (25%) Histology Melanoma 14 (50%) 5 Gy bid x 2-4 fx to one site RCC 12 (43%) Bladder 1 (3.5%) Sarcoma 1 (3.5%) IL-2 at 720,000 IU/kg q8 hr x 15 planned RT Site doses +/- TILs to start 2-24 hrs after RT Lung parenchyma 7 (25%) Lung hilum/mediastinum 5 (18%) Adrenal 3 (11%) Bone 4 (14%) IL-2 cycle 2 after 7-10 day break Soft tissue 5 (18%) Abdominal mass 3 (11%) Liver 1 (4%) J Immunother . 1991;12:265-71 PILOT STUDY OF RT AND IL2 • 5 patients received TIL – one PR in field. RT field Outside RT (%) field (%) Complete Response 1 (4) 0 (0) Partial Response 3 (11) 2 (7) • Why no benefit? Stable Disease 13 (46) 5 (18) Progressive Disease 8 (29) 20 (71) • RT dose too low? Inevaluable 3 (11) 1 (4) Overall Response (CR+PR) 4 (14) 2 (7) • RT field to large? J Immunother . 1991;12:265-71 PHASE I STUDY OF SBRT AND IL2 • Metastatic melanoma or RCC with at least 2009-2010 n=12 one lesion amenable to SBRT in the lung, Median Age (range), yrs 61 (51-65) mediastinum, or liver and at least one other Sex site not treated with SBRT. Male 10 (83%) Female 2 (17%) Histology 20 Gy x 1 fx to one site (2 fx in Melanoma 7 (58%) cohort 2 and 3 fx in cohort 3) RCC 5 (42%) SBRT Site Peripheral lung 5 (42%) IL-2 at 600,000 IU/kg q8 hr x 14 planned Central lung 2 (17%) doses to start 3 days after SBRT Mediastinum 1 (8%) Liver 4 (33%) SBRT site max diameter (range), cm 1.8 (0.5-6.1) IL-2 cycle 2 after 16 day break Re-image and repeat IL-2 course if regression Sci Transl Med . 2012;137:137ra74 PHASE I STUDY OF SBRT AND IL2 CT (%) PET (%) Complete Response 1 (8.4) 6 (50) Partial Response 7 (58.3) 2 (16.7) Stable Disease 1 (8.4) 1 (8.4) Progressive Disease 3 (25) 3 (25) Overall Response (CR+PR) 8 (66.7) 8 (66.7) Melanoma (n=7) CR 1 (14.3) 5 (71.4) PR 4 (57.1) 0 RCC (n=5) CR 0 (0) 1 (20) PR 3 (60) 2 (40) • ORR for melanoma 71% was > 16% historical for IL-2. • Of 8 responding patients – 6 maintained response median 480 days. Sci Transl Med . 2012;137:137ra74 PHASE I STUDY OF SBRT AND IL2 • Responders had a higher frequency of proliferating FOXP3 -, Ki67 + CD4 + TEM phenotype cells as well as CD8 + TEM phenotype at baseline and through day 15. Sci Transl Med . 2012;137:137ra74 RT AND IL2 • What will be the role of high dose IL-2 as other immunotherapy strategies evolve and play a more prominent role? Samples of Ongoing Clinical Trials of IL-2 and RT Primary Trial ID Accrual Goal Design Histology Endpoint Arm 1: High-Dose IL-2 alone NCT01416831 44 Metastatic Melanoma ORR (Phase II) Arm 2: High Dose IL-2 and SBRT (20 Gy x 1 or 20 Gy x 2) Arm 1: High-Dose IL-2 alone NCT01416831 84 Metastatic Melanoma ORR (Phase II) Arm 2: High Dose IL-2 and SBRT (20 Gy x 2) NCT01896271 26 High Dose IL-2 and SBRT (20 Gy x 1-3 fx) Metastatic Clear Cell RCC ORR (Phase II) C PRE-CLINICAL AND CLINICAL EVIDENCE Mφ DENDRITIC CELL (DC) CD8+ NK T cell Flt3-L TGF-β PD-L1 IL-6, IL-10 GM-CSF PRODUCTION Galactin-1 IDO DC MHC Loss RT DC CD8+ DC T cell CTLA-4 CTLA-4 PD-1 PD-1 CD8+ MDSC Treg T cell RT ABSCOPAL EFFECT IMMUNE MEDIATED • Metastatic mouse mammary carcinoma 67NR or A20 lymphoma -> injected s.c. into syngeneic mice in 2 sites -> treatment when primary tumor 100-150 mg 1. Control Control RT 2. DC growth factor Flt3-L Flt3-L 3. RT (2 Gy x 1 or 6 Gy x 1) to 1 o o 4. RT to 1 + Flt3-L RT + Flt3-L In nude mice -> no secondary tumor growth delay with Flt3-L + RT RT RT + Flt3-L Int J Radiation Oncology Biol Phys . 2004;58:862-70 RT + GM-CSF – PROOF OF PRICIPLE TRIAL • Stable or progressing metastatic solid 2003-2012 n=41 tumors with at least 3 distinct sites of Median Age (range), yrs 62 (54.5-69.5) measurable disease. Maintained on single Sex Male 8 (20%) agent chemotherapy or hormonal therapy. Female 33 (80%) Number of previous therapies RT 1 (0-3) 35 Gy in 10 fx to Chemotherapy 3 (2-4) one metastasis Number of measurable lesions Chest 2 (1-3) Abdomen 0 (0-0.5) GM-CSF 125 µg/m 2 daily Pelvis 0 (0-0) for 2 weeks starting week Any site 3 (2-4) two of RT Number of patients with lesions 3 lesions 21 (51%) 4-6 lesions 15 (37%) Course repeated to > 6 lesions 5 (12%) 2nd lesion starting d22 Lan cet Oncol. 2015;16:795-803 RT + GM-CSF – PROOF OF PRICIPLE TRIAL • Simon’s optimal two-stage design: Patients Not Assessable for best assessable abscopal response Patients could only be enrolled in stage for best 2 if at least one among the first 10 had abscopal PD/SD PR/CR an abscopal response.
Recommended publications
  • In Vivo Imaging of Tgfβ Signalling Components Using Positron
    REVIEWS Drug Discovery Today Volume 24, Number 12 December 2019 Reviews KEYNOTE REVIEW In vivo imaging of TGFb signalling components using positron emission tomography 1 1 2 Lonneke Rotteveel Lonneke Rotteveel , Alex J. Poot , Harm Jan Bogaard , received her MSc in drug 3 1 discovery and safety at the Peter ten Dijke , Adriaan A. Lammertsma and VU University in 1 Amsterdam. She is Albert D. Windhorst currently finishing her PhD at the VU University 1 Department of Radiology and Nuclear Medicine, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands Medical Center (VUmc) 2 under the supervision of A. Pulmonary Medicine, Institute for Cardiovascular Research, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands D. Windhorst and Adriaan A. Lammertsma. Her 3 research interest is on the development of positron Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, The emission tomography (PET) tracers that target Netherlands selectively the activin receptor-like kinase 5 in vitro and in vivo. Alex J. Poot obtained his The transforming growth factor b (TGFb) family of cytokines achieves PhD in medicinal chemistry homeostasis through a careful balance and crosstalk with complex from Utrecht University. As postdoctoral researcher at signalling pathways. Inappropriate activation or inhibition of this pathway the VUmc, Amsterdam, he and mutations in its components are related to diseases such as cancer, developed radiolabelled anticancer drugs for PET vascular diseases, and developmental disorders. Quantitative imaging of imaging. In 2014, he accepted a research expression levels of key regulators within this pathway using positron fellowship from Memorial Sloan Kettering Cancer 13 emission tomography (PET) can provide insights into the role of this Center, New York to develop C-labelled probes for tumour metabolism imaging with magnetic resonance in vivo pathway , providing information on underlying pathophysiological imaging (MRI).
    [Show full text]
  • Therapeutic PD-1 Pathway Blockade Augments with Other Modalities of Immunotherapy to Prevent Immune Decline in Ovarian Cancer
    Author Manuscript Published OnlineFirst on August 23, 2013; DOI: 10.1158/0008-5472.CAN-13-1550 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Therapeutic PD-1 Pathway Blockade Augments with other Modalities of Immunotherapy to Prevent Immune Decline in Ovarian Cancer Jaikumar Duraiswamy1, Gordon J. Freeman2 and George Coukos1,3* 1Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, PA, USA 2Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA 3Department of Oncology and Ludwig Center for Cancer Research, University Hospital of Lausanne, Switzerland Running Title: PD-1 blockade restores immune function in ovarian cancer *Correspondence: Dr. George Coukos MD, PhD Email: [email protected] Abbreviations: ID8, ovarian cancer model; TIL, tumor-infiltrating lymphocytes; PD-1, programmed death-1; PD-L, programmed death-1 ligands; TAM, tumor associated macrophages; DC, dendritic cells; MDSC, myeloid derived suppressor cells; Tregs, Foxp3+CD4+CD25+T cells; ID8-GVAX or ID8-FVAX, ID8 tumor cells transduced to express GM-CSF or Flt3-ligand Words in text (excluding references): 5493 Number of Figures: 7 Number of supplementary figures: 8 References: 56 1 Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 23, 2013; DOI: 10.1158/0008-5472.CAN-13-1550 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Abstract The tumor microenvironment mediates induction of the immunosuppressive programmed death-1 (PD-1) pathway, targeted interventions against which can help restore antitumor immunity.
    [Show full text]
  • Biological Therapy in Systemic Sclerosis
    Chapter 7 Biological Therapy in Systemic Sclerosis Joana Caetano, Susana Oliveira and JoanaJosé Delgado Caetano, Alves Susana Oliveira and AdditionalJosé Delgado information Alves is available at the end of the chapter Additional information is available at the end of the chapter http://dx.doi.org/10.5772/intechopen.69326 Abstract Systemic sclerosis is the autoimmune connective tissue disease with the highest morbid- ity and mortality, through the combination of inflammation, vasculopathy and fibrosis leading to severe internal organ involvement. Currently, there are no approved disease- modifying therapies, and treatment is based on organ-specific treatment and broad immu- nosuppression, with disappointing long-term results in most cases. Recent research has helped to improve knowledge of the pathogenesis of systemic sclerosis and to optimize treatment based on specific physiopathological targets, and a new era of biological agents in systemic sclerosis has now begun. Promising results are emerging from targeting spe- cific cytokine signalling, especially IL-6, and cellular subpopulations such as B cells, with anti-CD20 therapy, and T-cells, with inhibition of T-cell co-stimulation. Other approaches under evaluation are based on the modulation of profibrotic pathways by anti-TGF-β agents. In this chapter, we discuss the available evidence to support the use of each biologi- cal agent in systemic sclerosis based on data from basic and translational research and on results from clinical studies. Keywords: systemic sclerosis, biological therapy, cytokines, immune dysfunction, targeted treatment 1. Introduction Systemic sclerosis (SSc) is a rare multisystem connective tissue disease in which inflamma- tion, fibrosis, vasculopathy and autoimmunity are the principal features leading to diverse organ-based injuries [1].
    [Show full text]
  • Treatment of Diffuse Cutaneous Systemic Sclerosis with Hyperimmune Caprine Serum
    University College London (UCL) MD (Res) Degree Treatment of Diffuse Cutaneous Systemic Sclerosis with Hyperimmune Caprine Serum DR. NIAMH PATRICIA QUILLINAN 1 Declaration I, Niamh Patricia Quillinan, confirm that the work presented in this thesis is my own. Where information has been derived from other sources, I confirm that this has been indicated in the thesis. Signed: ___________________________________ Date: 17/03/2016 2 Abstract Systemic sclerosis (SSc) is a multisystem autoimmune rheumatic disorder with high morbidity and the highest case specific mortality of the rheumatic diseases. There is no currently approved unequivocally effective treatment for SSc and therefore there is a huge unmet medical need for novel and effective therapies. Hyperimmune caprine serum (HCS) is a goat serum extract derivative produced from goats vaccinated with a detergent-inactivated HIV viral lysate. It contains caprine immunoglobulins and small molecular weight proteins as well as a CRH, α-2 macroglobulin (α-2M) and lipoprotein-related peptide-1 complex. In this thesis we explore the hypothesis that hyperimmune caprine serum improves skin and other measures of disease severity in established dcSSc by modulating immunological function that determines persistence of clinical disease. This hypothesis is explored through 1) a prospective clinical trial, 2) long-term clinical use and 3) detailed assessment of serum growth factors and cytokines, as well as established and exploratory markers of disease. The primary objective of the clinical trial was to explore safety and tolerability of HCS in established diffuse cutaneous systemic sclerosis (dcSSc). Secondary objectives included assessment of potential efficacy and biological activity and exploration of candidate biomarkers. There were no safety concerns and frequency of adverse events was not different between HCS and placebo group.
    [Show full text]
  • Characterization of the CCL21-Mediated Melanoma-Specific Immune Responses and in Situ Melanoma Eradication
    1755 Characterization of the CCL21-mediated melanoma-specific immune responses and in situ melanoma eradication Laura Novak, Olga Igoucheva, Stephanie Cho, Introduction and Vitali Alexeev The incidence of cutaneous malignant melanoma is increasing at a faster rate than any other cancer, both Department of Dermatology and Cutaneous Biology, Jefferson Medical College, Thomas Jefferson University, Philadelphia, globally and in the United States (1). Immunotherapy has Pennsylvania emerged as one of the most promising treatments of this melanocytic malignancy. Various vaccine therapies to achieve immune-based melanoma rejection have been Abstract successfully tested in preclinical animal models and several Previous studies have shown that secondary lymphoid clinical trials. However, clinical studies have shown that chemokine, CCL21, can be used for modulation of tumor- existing therapeutic approaches are generally ineffective or specific immune responses. Here, using B16F0 melanoma prohibitively expensive. cells stably expressing CCL21 under the control of Recently, chemokines have attracted considerable interest cytomegalovirus and ubiquitin promoters, we showed that within the field of tumor immunotherapy. Chemokines are CCL21-activated immune responses depend on the small proteins that are involved in immune and inflamma- amount of melanoma-derived chemokine, which, in turn, tory responses. One of them, secondary lymphoid chemo- depends on the strength of the promoter. We showed that kine CCL21 (also known as SLC or C6kine), is prominently ubiquitin promoter–driven expression of CCL21 enabled expressed in high endothelial venules and within T-cell + À + massive infiltration of tumors with CD4 CD25 , CD8 zones of secondary lymphoid organs, where it recruits + T lymphocytes, and CD11c dendritic cells, and conse- naive T cells and antigen-presenting cell (APC), such as quent activation of cellular and humoral immune dendritic cell, via a mechanism known as chemoattraction.
    [Show full text]
  • Clinical Research Newsletter for Colleagues in the Community
    Clinical Research Newsletter for Colleagues in the Community Welcome to the Winter 2017 issue of the Stanford Cancer surgery, robotic surgery, stereotactic radiosurgery such as Institute Clinical Research Newsletter. This quarterly CyberKnife®, microvascular reconstruction, intraoperative publication is designed to inform our colleagues in the radiation therapy (IORT), along with new chemotherapy trials. medical community, and especially physicians who are The Stanford Cancer Institute Neuro-Oncology Program considering treatment options for their patients with offers Phase I through III trials as well as multidisciplinary, cancer, about current clinical trials available at the Stanford collaborative evaluation and treatment of patients with tumors Cancer Institute, a National Cancer Institute designated of the nervous system. This includes, but is not restricted Comprehensive Cancer Center. Many of these trials provide to; brain metastases, leptomeningeal cancer, glioblastomas access to novel therapies including new “targeted” agents, and less aggressive gliomas, benign brain and spinal tumors, often not available in the community. and base of brain neoplasms including pituitary disorders. As leaders of the Stanford Head and Neck Cancer Care Clinical trials have focused on vaccine therapy, antibody Program, we are delighted to introduce this edition of the therapy, novel chemotherapy agents, radiation sensitizers, newsletter, as it focuses on our Neuro-Oncology, Thoracic novel radiation therapy, and radiosurgery techniques. Oncology, and Developmental Therapeutics programs. The Developmental Therapeutics Program conducts Each of these programs offers cutting-edge clinical trials pharmacokinetic and pharmacodynamic driven first-in- for patients with tumors that can be challenging to treat human trials tailored to make early, informed decisions with current routine care. Weekly multidisciplinary tumor regarding the suitability of novel molecular agents for boards are available for each program.
    [Show full text]
  • (12) Patent Application Publication (10) Pub. No.: US 2017/0172932 A1 Peyman (43) Pub
    US 20170172932A1 (19) United States (12) Patent Application Publication (10) Pub. No.: US 2017/0172932 A1 Peyman (43) Pub. Date: Jun. 22, 2017 (54) EARLY CANCER DETECTION AND A 6LX 39/395 (2006.01) ENHANCED IMMUNOTHERAPY A61R 4I/00 (2006.01) (52) U.S. Cl. (71) Applicant: Gholam A. Peyman, Sun City, AZ CPC .......... A61K 9/50 (2013.01); A61K 39/39558 (US) (2013.01); A61K 4I/0052 (2013.01); A61 K 48/00 (2013.01); A61K 35/17 (2013.01); A61 K (72) Inventor: sham A. Peyman, Sun City, AZ 35/15 (2013.01); A61K 2035/124 (2013.01) (21) Appl. No.: 15/143,981 (57) ABSTRACT (22) Filed: May 2, 2016 A method of therapy for a tumor or other pathology by administering a combination of thermotherapy and immu Related U.S. Application Data notherapy optionally combined with gene delivery. The combination therapy beneficially treats the tumor and pre (63) Continuation-in-part of application No. 14/976,321, vents tumor recurrence, either locally or at a different site, by filed on Dec. 21, 2015. boosting the patient’s immune response both at the time or original therapy and/or for later therapy. With respect to Publication Classification gene delivery, the inventive method may be used in cancer (51) Int. Cl. therapy, but is not limited to such use; it will be appreciated A 6LX 9/50 (2006.01) that the inventive method may be used for gene delivery in A6 IK 35/5 (2006.01) general. The controlled and precise application of thermal A6 IK 4.8/00 (2006.01) energy enhances gene transfer to any cell, whether the cell A 6LX 35/7 (2006.01) is a neoplastic cell, a pre-neoplastic cell, or a normal cell.
    [Show full text]
  • Recommended INN: List 63
    WHO Drug Information, Vol. 24, No. 1, 2010 Recommended INN: List 63 International Nonproprietary Names for Pharmaceutical Substances (INN) RECOMMENDED International Nonproprietary Names: List 63 Notice is hereby given that, in accordance with paragraph 7 of the Procedure for the Selection of Recommended International Nonproprietary Names for Pharmaceutical Substances [Off. Rec. Wld Health Org., 1955, 60, 3 (Resolution EB15.R7); 1969, 173, 10 (Resolution EB43.R9); Resolution EB115.R4 (EB115/2005/ERC/1)], the following names are selected as Recommended International Nonproprietary Names. The inclusion of a name in the lists of Recommended International Nonproprietary Names does not imply any recommendation of the use of the substance in medicine or pharmacy. Lists of Proposed (1–101) and Recommended (1–62) International Nonproprietary Names can be found in Cumulative List No. 13, 2009 (available in CD-ROM only). Dénominations communes internationales des Substances pharmaceutiques (DCI) Dénominations communes internationales RECOMMANDÉES: Liste 63 Il est notifié que, conformément aux dispositions du paragraphe 7 de la Procédure à suivre en vue du choix de Dénominations communes internationales recommandées pour les Substances pharmaceutiques [Actes off. Org. mond. Santé, 1955, 60, 3 (résolution EB15.R7); 1969, 173, 10 (résolution EB43.R9); résolution EB115.R4 (EB115/2005/ERC/1)] les dénominations ci-dessous sont choisies par l’Organisation mondiale de la Santé en tant que dénominations communes internationales recommandées. L’inclusion d’une dénomination dans les listes de DCI recommandées n’implique aucune recommandation en vue de l’utilisation de la substance correspondante en médecine ou en pharmacie. On trouvera d’autres listes de Dénominations communes internationales proposées (1–101) et recommandées (1–62) dans la Liste récapitulative No.
    [Show full text]
  • Fresolimumab Suppl Methods
    Supplementary Figure 1. Fresolimumab treated patients show decreased biomarkers and skin scores. Graphs show thrombopondin-1 (THBS1, panels a and b) and cartilage oligomeric protein (COMP, panels c and d) gene expression, and MRSS (panels e and f) from study patients in group 1, receiving two doses of 1 mg/kg fresolimumab, (a, c, and e) and group 2, receiving one dose of 5 mg/kg fresolimumab (b, d and f). Line graphs show changes in individual patients over time. A B C Supplementary Figure 2: Biomarker gene expression correlates with the local skin score. The local forearm skin score correlates highly with the MRSS (panel a), as well as THBS1 (panel b) and COMP (panel c) gene expression. Supplementary Figure 3. Autoantibody levels measured at baseline and 11 weeks after fresolimumab treatment. Anti-RNA polymerase III levels for anti-RNA polymerase III- positive patients are shown in blue. Anti-Scl70 levels for anti-Scl70-positive patients are shown in red. Autoantibody levels in two of the four anti-Scl70-positive patients lie on top of the other two. Supplementary Figure 4. Disease duration compared to the change in MRSS 7 weeks after fresolimumab treatment. The change in MRSS after fresolimumab treatment showed no correlation with the disease duration (r2 = 0.00063) Supplementary Figure 5A. Immunohistochemical Supplementary Figure 5B. Skin collagen staining of plasminogen activator protein-1 (PAI- thickness. Based on trichrome staining, dermal 1/serpine1). Each patient is represented by a thickness was measured from the dermal-epidermal different color/style of marker. The median of the junction to subcutaneous fat.
    [Show full text]
  • Epithelial–Mesenchymal Transition in Crohn's Disease
    REVIEW Epithelial–mesenchymal transition in Crohn’s disease H Jiang1, J Shen1 and Z Ran1 Crohn’s disease (CD) is often accompanied by the complications of intestinal strictures and fistulas. These complications remain obstacles in CD treatment. In recent years, the importance of epithelial–mesenchymal transition in the pathogenesis of CD-associated fistulas and intestinal fibrosis has become apparent. Epithelial–mesenchymal transition refers to a dynamic change, wherein epithelial cells lose their polarity and adherence and acquire migratory function and fibroblast features. During formation of CD-associated fistulas, intestinal epithelial cells dislocate from the basement membrane and migrate to the lining of the fistula tracts, where they convert into transitional cells as a compensatory response under the insufficient wound healing condition. In CD-associated intestinal fibrosis, epithelial–mesenchymal transition may serve as a source of new fibroblasts and consequently lead to overproduction of extracellular matrix. In this review, we present current knowledge of epithelial–mesenchymal transition and its role in the pathogenesis of CD in order to highlight new therapy targets for the associated complications. INTRODUCTION and fistula formation in CD.6,7 EMT was first identified in the Therapeutic strategies for Crohn’s disease (CD) have progressed 1960s from the pioneering work of Elizabeth Hay, who remarkably in the past decade.1 From 1991 to 2014, the rate of described transformation of epithelial cells to mesenchymal hospitalization and surgical intervention for CD decreased; cells in the chick primitive streak during embryogenesis.8 however, despite the use of advanced drugs such as immuno- Generally, in EMT, epithelial cells demonstrate plasticity and modulators and biologics, the progression to complicated disease lose epithelial markers like E-cadherin and catenins.
    [Show full text]
  • Cancer Vaccines in Ovarian Cancer: How Can We Improve?
    biomedicines Review Cancer Vaccines in Ovarian Cancer: How Can We Improve? Silvia Martin Lluesma 1, Anita Wolfer 2, Alexandre Harari 1 and Lana E. Kandalaft 1,3,* 1 Center of Experimental Therapeutics, Ludwig Center for Cancer Res, Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland; [email protected] (S.M.L.); [email protected] (A.H.) 2 Department of Oncology, University of Lausanne, Lausanne 1011, Switzerland; [email protected] 3 Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, PA 19104, USA * Correspondence: [email protected]; Tel.: +41-(0)21-314-7823 Academic Editor: Ghaith Bakdash Received: 21 March 2016; Accepted: 19 April 2016; Published: 3 May 2016 Abstract: Epithelial ovarian cancer (EOC) is one important cause of gynecologic cancer-related death. Currently, the mainstay of ovarian cancer treatment consists of cytoreductive surgery and platinum-based chemotherapy (introduced 30 years ago) but, as the disease is usually diagnosed at an advanced stage, its prognosis remains very poor. Clearly, there is a critical need for new treatment options, and immunotherapy is one attractive alternative. Prophylactic vaccines for prevention of infectious diseases have led to major achievements, yet therapeutic cancer vaccines have shown consistently low efficacy in the past. However, as they are associated with minimal side effects or invasive procedures, efforts directed to improve their efficacy are being deployed, with Dendritic Cell (DC) vaccination strategies standing as one of the more promising options. On the other hand, recent advances in our understanding of immunological mechanisms have led to the development of successful strategies for the treatment of different cancers, such as immune checkpoint blockade strategies.
    [Show full text]
  • Targeting TGF-Β Signaling for Therapeutic Gain
    Downloaded from http://cshperspectives.cshlp.org/ on September 25, 2021 - Published by Cold Spring Harbor Laboratory Press Targeting TGF-b Signaling for Therapeutic Gain Rosemary J. Akhurst Department of Anatomy and Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94158-9001 Correspondence: [email protected] Transforming growth factor bs (TGF-bs) are closely related ligands that have pleiotropic activity on most cell types of the body. They act through common heterotetrameric TGF-b type II and type I transmembrane dual specificity kinase receptor complexes, and the outcome of signaling is context-dependent. In normal tissue, they serve a role in maintaining homeostasis. In many diseased states, particularly fibrosis and cancer, TGF-b ligands are overexpressed and the outcome of signaling is diverted toward disease progression. There has therefore been a concerted effort to develop drugs that block TGF-b signaling for therapeutic benefit. This review will cover the basics of TGF-b signaling and its biological activities relevant to oncology, present a summary of pharmacological TGF-b blockade strategies, and give an update on preclinical and clinical trials for TGF-b blockade in a variety of solid tumor types. he three transforming growth factor bs titude of TGF-b-induced tumor promoting ef- T(TGF-bs), TGF-b1, -b2, and -b3, are closely fects modulate the tumor cells directly through related ligands that act on most cells types of the enhancement of tumor cell invasion and metas- body and have pleiotropic activities. Expression tasis, and induction and maintenance of cells of TGF-b1 is activated by tissue perturbations with tumor initiating properties, sometimes that induce cellular stress, such as cell prolifera- termed cancer stem-like cells (CSCs).
    [Show full text]