Search Results for: ojas stem cells

Umoja Biopharma Presents Data on its Engineered Induced Pluripotent Stem Cell Platform at the 2022 International Society for Stem Cell Research Annual…

Posted: June 22, 2022 at 2:09 am

News and research before you hear about it on CNBC and others. Claim your 1-week free trial to StreetInsider Premium here.

SEATTLE, June 16, 2022 (GLOBE NEWSWIRE) -- Umoja Biopharma, Inc., an immuno-oncology company pioneering off-the-shelf, integrated therapeutics that reprogram immune cells in vivo to treat patients with solid and hematologic malignancies, announced today that it will have a poster presentation at the 2022 International Society for Stem Cell Research (ISSCR) Annual Meeting, to be held June 15-18, 2022 in San Francisco, California.

On Wednesday, June 15th, Principal Scientist & iPSC Team Lead, Teisha Rowland, Ph.D., will give a poster presentation titled, A Synthetic Cytokine Receptor Platform for Producing Cytotoxic Innate Lymphocytes as Off-the-Shelf Cancer Therapeutics. The presentation will discuss Umojas engineered induced pluripotent stem cell (iPSC) platform, that incorporates the synthetic cytokine receptor system rapamycin-activated cytokine receptor (RACR) platform. Umojas engineered iPSCs that are modified to express RACR, called RACR-induced cytotoxic innate lymphoid (iCIL) cells, drive differentiation and expansion of the cells while eliminating the need for expensive cytokines and other raw materials. The RACR platform has the potential to enable cytokine-free manufacturing and engraftment of the engineered cells in the patient without the need for toxic lymphodepletion.

Despite the advances chimeric antigen receptor T cell therapies have provided to the oncology space, we continue to battle significant challenges that these therapies cannot address, like limited expansion capacity and scalability, manufacturing complexity, variability among patients, and the need for toxic chemotherapy administration to combat patients anti-allograft response, said Andy Scharenberg, M.D., co-founder and Chief Executive Officer of Umoja. We are developing an engineered iPSC platform, including the RACR platform, to address these challenges by enabling a scalable, virtually unlimited, and simplified manufacturing of engineered, cancer-fighting cytotoxic innate lymphocytes.

About Umoja Biopharma

Umoja Biopharma, Inc. is an early clinical-stage company advancing an entirely new approach to immunotherapy. Umoja Biopharma, Inc. is a transformative multi-platform immuno-oncology company founded with the goal of creating curative treatments for solid and hematological malignancies by reprogramming immune cells in vivo to target and fight cancer. Founded based on pioneering work performed at Seattle Childrens Research Institute and Purdue University, Umojas novel approach is powered by integrated cellular immunotherapy technologies including the VivoVec in vivo delivery platform, the RACR/CAR in vivo cell expansion/control platform, and the TumorTag targeting platform. Designed from the ground up to work together, these platforms are being developed to create and harness a powerful immune response in the body to directly, safely, and controllably attack cancer. Umoja believes that its approach can provide broader access to the most advanced immunotherapies and enable more patients to live better, fuller lives. To learn more, visit http://umoja-biopharma.com/.

About RACR

CAR T cells generated by the body with VivoVec can be expanded and sustained with the rapamycin activated cytokine receptor (RACR) system, an engineered signaling system designed to improve chimeric antigen receptor (CAR) T cell persistence and produce durable anti-tumor responses. The RACR/CAR payload is integrated into the genomic DNA of a patients T cells. Rapamycin activates the RACR system resulting in preferential expansion and survival of cancer-fighting T cells. The RACR technology enables a patients cells to expand in a manner that resembles a natural immune response that does not require lymphodepletion, promoting durable T cell engraftment. RACR/CAR technology can also be used to enhanceex vivomanufacturing in support of more traditional autologous or allogeneic cell therapy manufacturing processes. To learn more about Umojas RACR platform please visit https://www.umoja-biopharma.com/platforms/

Media Contact:Darren Opland, Ph.D.LifeSci Communications[emailprotected]

Follow this link:
Umoja Biopharma Presents Data on its Engineered Induced Pluripotent Stem Cell Platform at the 2022 International Society for Stem Cell Research Annual...

Posted in Stem Cell Treatments | Comments Off on Umoja Biopharma Presents Data on its Engineered Induced Pluripotent Stem Cell Platform at the 2022 International Society for Stem Cell Research Annual…

Stemcell treatment for hair and skin, Autologous Adipose Stem Cell Treatment – Video

Posted: December 16, 2013 at 4:45 am


Stemcell treatment for hair and skin, Autologous Adipose Stem Cell Treatment
Through the history of stem cell therapy and stem cell research, animal stem cells have been used, human embryonic stem cells, and now research has led us to...

By: Ojas Aesthetic

See original here:
Stemcell treatment for hair and skin, Autologous Adipose Stem Cell Treatment - Video

Posted in Cell Therapy, Stem Cell Therapy | Comments Off on Stemcell treatment for hair and skin, Autologous Adipose Stem Cell Treatment – Video

Ojas Stem Cell – Video

Posted: December 13, 2012 at 12:44 pm


Ojas Stem Cell
We use YOUR own stem cells because they are safer and more effectiveFrom:ojas aestheticViews:0 0ratingsTime:02:23More inHowto Style

Read the original here:
Ojas Stem Cell - Video

Posted in Stem Cell Videos | Comments Off on Ojas Stem Cell – Video

Diagnosis and treatment of the alpha-Gal syndrome | JAA – Dove Medical Press

Posted: July 19, 2022 at 2:27 am

Introduction

Galactose--1,3-galactose (-Gal), an oligosaccharide that structurally resembles blood group B antigen, is present in both glycoproteins and glycolipids from non-catarrhine mammalian muscle cells and secretions.1,2 Old World monkeys, apes and humans evolved with the inability to synthesize -Gal epitopes and, therefore, produce natural anti--Gal antibodies to control pathogen infection.3 This carbohydrate epitope is the causal agent of the -Gal syndrome (AGS), a pathognomonic immunoglobulin E (IgE)-mediated delayed anaphylaxis in mammalian meat (eg pork, beef or lamb) or dairy products 3 to 6 hours post-consumption.47 Recently, an allergic cross reaction to flounder roe in patients suffering from AGS has been reported.8 The other clinical presentations of AGS comprise immediate hypersensitivity to -Gal-containing drugs, firstly discovered using the monoclonal antibody cetuximab in anticancer therapy.6,9 There is growing evidence of allergic reactions caused by the -Gal present in mammalian substances such as gelatin, glycerin, lactic acid and magnesium stearate used in the preparation process of several medications,9,10 such as gelatin-containing products (vaccines and volume colloids), mammalian serum-based antivenom and even various analgesics and nonsteroidal anti-inflammatory drugs (NSAIDs).2,6,11,12 The IgE initial sensitization is caused by hard-bodied tick bites from different species according to geographic location and is attributed to -Gal-containing tick salivary glycoproteins, but also other tick salivary biomolecules without -Gal modifications such as prostaglandin E2 (PGE2).1317 The mechanisms behind tick -Gal induction of sensitization are still unknown, but besides the -Gal moiety, tick sialome components may play an important role in the chained immune reaction activation (Figure 1).18,19 Tick species such as Ixodes ricinus in Europe, Amblyomma americanum in North America, Haemaphysalis longicornis in Asia and Ixodes holocyclus in Australia are linked to AGS,20 currently considered an emergent life-threatening allergy in tick endemic areas worldwide.2123 However, not all individuals bitten by ticks or those that carry elevated specific IgE (sIgE) against -Gal develop AGS, in fact, the majority only produce sIgE against it.19 Symptomatic individuals typically show delayed pruritus, urticaria (acute or recurrent), angioedema, anaphylaxis, malaise or gut-related symptoms such as abdominal pain, vomits and diarrhea.22,24,25 Anaphylaxis has been triggered in up to 60% of AGS cases and can be fatal if it is not treated promptly.2628 Clinical features reported tend to be restricted to gastrointestinal complaints, hampering the suspicion of an allergic etiology.29 Nevertheless, clinical observations in AGS patients are widely variable, showing proof of individual sensitivity.5 Augmenting factors, also called cofactors, such as exercise and alcohol intake, have been reported to play an important role in modulation of this individual susceptibility between patients.30 The medical history is of importance in these cases and details like meat-associated delayed allergic reactions and tick bite-exposure represent crucial factors for uncovering AGS, which otherwise can be misdiagnosed as idiopathic anaphylaxis or chronic spontaneous angioedema.7,30,31 Risk factors for developing sIgE to -Gal are related to the probability of individual tick bite-exposure in certain environmental conditions, including practice of outdoor activities (eg, hunting or hiking), living in rural areas, pet-ownership, and certain jobs such as forest service employees.3235 The sIgE values tend to increase according to the number of tick bites per year and on how recent those bites are.34,36 Moreover, individuals that do not have type B or AB blood group may have a higher risk of developing AGS, as blood group B antigen, similar to -Gal, creates tolerance to this epitope.37

Figure 1 Alpha-Gal syndrome (AGS). (A) Sensitization after several tick bites. Tick saliva contains glycoproteins, glycolipids with -Gal epitopes and other unknown salivary biomolecules that could be involved in the pathology of AGS. The glycan -Gal is presented to T helper 2 (Th2) cells through antigen-presenting cells (APCs) as dendritic cells, macrophages or even B cells. Once T cells are activated, B cells are leading to produce IgE against -Gal (anti -Gal-IgE) in an enriched interleukin environment and potentiate IgE production in plasma cells. Free IgE are now available to interact and bind to IgE receptors present in basophils and mast cells. (B) Allergic Reaction. When AGS patients ingest mammalian meat containing -Gal bound to proteins or lipids, these molecules expressing the allergen epitope are absorbed and incorporated to lipid/protein macromolecules during digestion (chylomicrons, lipoproteins), which will be processed and transport through protein or lipid metabolism to systemic circulation and peripheral tissues. About 36 hours post-consumption, IgE-mediated and coated effectors will recognize the allergen, leading to degranulation of basophils and mast cells and promoting a systemic delayed allergic reaction. AGS can also comprise an immediate anaphylactic reaction, triggered using -Gal containing drugs, administered via parenteral due to therapeutic reasons.

On the other hand, immune response to -Gal has been studied for the control and prevention of diseases, exhibiting a protective role in human evolution catastrophic selection. The incidence of several infectious diseases caused by -Gal containing-pathogens such as malaria or tuberculosis might be positively correlated with the frequency of the specific blood type B, and then, with a reduced immune response to -Gal. However, this fact has been associated with a lower prevalence of food allergies related to anti--Gal IgE antibodies.38 In addition, it has been recently reported a positive correlation between anti--Gal IgM antibodies and the incidence of Plasmodium falciparum infection, decreasing its transmission.39 Despite the fact that uncontrolled levels of anti--Gal antibodies could compromise health in AGS patients, these findings suggest that anti--Gal antibodies (IgE or IgM) might protect against parasites containing -Gal on their surface. Interestingly, anti--Gal antibodies have also been studied not only in vector-borne diseases but also in emerging virus infections. Recently, it has been reported that anti--Gal antibody levels negatively correlate not only with SARS-CoV-2 infection but also with COVID-19 symptomatology severity.40

Over 10 years have passed since the discovery of AGS,41 but many questions remain unclear that still need to be elucidated, especially those related to the diagnostic and therapeutical approaches used for this syndrome. The aim of the present review article is to outline current diagnostic methods used for AGS and potentially future diagnostic tools, combined with the most recent forms of treatment/management of this syndrome. Furthermore, innovative topics such as current research methods and future treatment and preventive strategies are also discussed.

The AGS is an allergic disorder that challenges clinical diagnosis due to inapparent presentation and delayed reactions.19,42 Like any other allergic disease, diagnosis relies on a well-detailed medical history in order to reach an accurate evaluation and prognostic of individual signs and symptoms.25 Diagnostic techniques for this syndrome are not specific and/or represent a risk for the patients health, whereas more precise methods still show limitations to its use.43 As discussed here, it is important to address the diagnostic tests more commonly used and methods that could potentially be employed in the future (Figure 2) for the challenging and complex allergy that involves the pathology of AGS.

Figure 2 Conventional and next generation methods for the diagnosis of the alpha-Gal syndrome (AGS).

SPTs remains a useful diagnostic tool for several food allergies.44 Conversely, conducting this test for AGS diagnosis using conventional and commercially available mammalian meat extracts (beef, pork or lamb) lacks sensitivity, yielding low-reactive results (24 mm wheals), which may lead to misdiagnosis and incorrect patient management.45 Alternatively, cancer drug cetuximab can potentially be used as a sensitivity agent due to its high capacity to induce a strong skin reactivity in AGS patients, mainly caused by the larger amount of -Gal epitopes exposed to the surface.46,47 Robust reactions also occur when mammalian meat extract is used, although this is not a feasible option for daily practice.16 Meat-derived gelatin from porcine or bovine, sometimes forming colloids, has also been used in allergic reaction diagnostics. Furthermore, it is important to consider that, although rare, the use of high-sensitivity components in SPT could potentially trigger a fatal anaphylactic shock reaction.48,49 Less commonly, intradermal testing (IDT) can also be used as a standardized methodology to evaluate skin reactions. As described by the SPT method, 20 minutes after allergen intradermal injection (around 0.1 mg/mL), swelling, redness and wheals are observed in the area of injection.50 Nevertheless, IDT is more likely to induce systemic anaphylactic reactions when compared to SPT.44 Overall, the clinical utility of SPT remains doubtful as no food allergen fits flawlessly in this diagnostic technique, conveying on several limitations and therefore making it not fit for a primary approach diagnostic tool.

For the diagnosis of a food allergy (FA), OFC is the gold standard technique, offering further information regarding food tolerability and threshold of responsiveness.51 This method could be useful for discriminating AGS diagnosis from -Gal sensitization if it did not convey to a risk of fatal or near-fatal delayed anaphylactic reactions.7,52 Therefore, this challenge can only be performed in specialized allergic centers, requiring long patient observation periods.7,53 Besides risk, OFC is not established as a standardized procedure for AGS and exposure reaction presents a high variability between patients. In fact, some patients may require the presence of cofactors in order to react, while others only respond to a particular type of red meat.54 Nevertheless, this method, tested also in combination with cofactors, is essential in patients in which drugs-containing -Gal must be given for therapeutical reasons due to the clinically relevant information that it provides.55 Cofactors, such as acetylsalicylic acid (ASA) or alcohol, are well-known amplifiers of -Gal reactions.6 Due to the wide difference between symptomatology of AGS patients, sensitivity to -Gal can be truly variable.30 However, it has been observed that pork kidney intake, and no other product like muscle meat, even in the coadministration of cofactors, is a key element to raise AGS symptoms. This difference might be explained due to the higher number of -Gal epitopes present in pork or beef kidney in comparison to other meats/innards.56,57

Currently, serum anti-Gal IgE levels measured using an immune-enzymatic assay (bovine thyroglobulin-conjugated ImmunoCAP) is the confirmatory diagnostic method used for AGS diagnosis when medical history matches with this disease.5860 Nonetheless, it remains unclear the clinical relevance of positive testing for anti-Gal IgE using a cut-off value of 0.35 kU/L (where 1 kU/L = 1 IU/mL = 2.4 ng/mL).33,42,46,52,61 While Mabelane et al54 state that 5.5 kU/L is the cut-off point for clinically significant AGS, other studies reveal that there are no strict criteria regarding anti-Gal IgE levels as an allergic symptomatology predictor.25,62 One thing is clear, though, is that levels of specific IgE are not a useful biomarker for predicting the severity of allergic reactions, as AGS patients experiencing anaphylactic reactions may maintain IgE levels overtime or even in rare occasions with anti-Gal IgE negative results.25,63 Another issue with the anti-Gal IgE diagnostic assay is that it can lead to false-positive results in those individuals where Gal IgE sensitization may also be related to bee and wasp stings, parasitism, atopy or cat ownership, creating cases where these antibodies do not match the clinically pathognomonic history of AGS.25,42,62,64,65 For example, in a clinical study carried out in southern Germany, among 300 hunters with a 19.3% of IgE--Gal prevalence (58 individuals positive for cut-off value of 0.35 IU/mL), only 1.67% (5 individuals of the initial cohort) had allergic reactions to mammalian meat.33 Moreover, serum levels of IgE to Gal tend to drop when patients do not experience recurrent tick bites, but again, the rate of declination between individuals is widely variable, being therefore recommended to repeat testing every 8 to 12 months.25,45 Nevertheless, due to AGS non-related therapeutical reasons, sometimes the measurement of anti-Gal IgE levels may be needed to detect Gal sensitization and therefore prevent drug-induced anaphylaxis.60 In summary, anti-Gal IgE levels may be useful for AGS diagnosis, but clinical symptomatology and disease severity cannot be evaluated exclusively through this parameter, requiring complementation with other diagnostic techniques.

Over the years, research studies allowed to recognize distinct improved biomarkers that provide a more accurate diagnosis of food allergies.66 This led the way to the use of the in vitro functional assay BAT, a flow-cytometry-based technique that quantifies the expression of activation membrane markers, namely, CD63 and CD203c, in order to analyze basophil degranulation when triggered by a specific allergen.6668 In the research setting and specialized referral centers, BAT is being used as a diagnostic clarifier, allowing to distinguish between merely asymptomatic sensitized individuals and patients suffering from AGS.42,53,69 This newly emerging method offers good sensitivity and specificity, but presents several practical and logistical issues for implementation in clinical practice.43,70 First, blood must be processed within 24 hours after being collected in order to guarantee that basophil viability and reactivity are preserved.6,71 Second, questions regarding reproducibility and cost must be addressed before BAT implementation in practicing allergists.72 Unfortunately, methodology, concentration and markers are not standardized between laboratories in order to allow result comparison and test validation.73 Furthermore, standardization between systems and instruments required for accreditation (eg, EuroFlow Standard Operating Procedures) is missing, reducing BAT availability.74,75 This technique also lacks an established proficiency testing by regulatory entities. The current European Directive 98/79/EC on in vitro diagnostic medical devices76 will be replaced in 2022 by the new Regulation (EU) 2017/746 and introduce major changes in the sector, aiming for a smooth functioning of the internal market.77 In sum, efforts should be made to convey on transforming BAT into an on-hand tool for clinicians, due to the benefits it presents on risk allergy stratification, precise decision-making for -Gal sensitized patients who lack medical evidence and selection of the correct doses for OFC in AGS-suffering individuals.53,72

Another in vitro assay executed by flow cytometry is the MAT, a technique that measures CD63, a membrane activation marker that increases when mast cells (MCs) degranulate. This phenomenon occurs when MCs are triggered by allergen-sIgE antibodies.66,78 MAT presents high sensitivity and leads to a dose-dependent response to allergens,79 making it a potential and attractive complementary candidate for AGS diagnosis. Furthermore, MAT seems to overcome BATs major limitations. First, the use of MCs rather than basophils appears to be more suitable for allergy diagnosis due to the well-recognized effector function of MCs in comparison to the mere regulatory activity of basophils.80 Secondly, serum samples can be frozen prior to their use, as MAT does not require fresh viable cells, facilitating logistics and sample shipment if required.81 The MCs line can be activated directly through mas-related G protein-coupled receptor X2 (MGPRX2) with simultaneous analysis of positive and negative populations for this receptor. Herein, MC degranulation can be studied via upregulation of specific degranulation markers, such as CD63. Most common MRGPRX2-expressing cell lines used in combination with CD63 detection by flow cytometry are LAD-2 cells derived from human CD34+ cells.82,83 However, there is still a long way to go from standardization to validation to obtain a fully functional MAT assay, as currently this technique is particularly time-consuming and several key issues still persist regarding heterogeneity of MCs.70,84 Although MAT test is still under validation for clinical application, it represents a promising diagnostic approach, particularly as a confirmatory test when conventional methods generate ambiguous results.66,82

Another emerging diagnostic test in the FA field is the HR assay, a standardized test based on fluorescence intensity that measures the amount of histamine released by activated basophils.85 Although even further studies are required to support standard results, this technique could potentially be used for AGS diagnosis since basophil reactivity was found to be higher in these patients when compared to -Gal sensitized individuals.10 Indeed, this method displays similar high sensitivity and specificity values when compared to the BAT test,86 but further studies are required to support this result, especially involving AGS-suffering patients.

Collective characterization and quantitation of biomolecules known as omics technologies such as metabolomics, metagenomics, proteomics and transcriptomics could advance knowledge of the immune response in AGS and its molecular drivers, enabling the identification of biotargets for molecular diagnosis of this global impact disease.10,87 Not only the identification of host biomarkers and host-derived immune response mechanisms but also tick-derived biomolecules are important for the development of new diagnostic tools.52 Proteins present in tick sialome, especially highly conserved across tick species, could potentially serve as diagnostic antigens.47 A recent study by Villar et al88 identified by proteomics analysis of tick sialome and alphagalactome the 14-3-3 family chaperone that could possibly constitute a future diagnostic disease biomarker. The study identified that 14-3-3 family chaperone and other proteins were recognized by IgE in sera from AGS patients.88 Therefore, they proposed that these proteins may potentially be involved in the AGS and other disorders with the possibility of mediating protective immune mechanisms against tick infestations and pathogen infection.88 Nevertheless, there are also tick salivary gland proteins with non--Gal modifications that could probably be used to develop ELISA tests for antibody quantification as a complementary diagnostic method for AGS.47,52,89

Artificial intelligence (AI) and machine learning are considered to be powerful diagnostic assistance tools, which in the future could revolutionize the healthcare system providing an accurate and custom-based diagnosis.90 MBR algorithms aim to create a clinical diagnosis or decision-making model that utilize hybrid reasoning and data-driven AI in order to obtain high diagnostic yields by combining the integrative medicine concept.91 For puzzling and complex diseases, such as AGS, in which diagnostic tools lacking standardization and cofactors are also involved, the use of this integrative diagnostic technique could represent the best fitting method. The use of this methodology has been proposed by de la Fuente et al52 for improving AGS diagnosis, considering together clinical symptoms, risk factors and anti--Gal sIgE levels. They also proposed that the machine learning algorithm could be transformed into a code for a software creation with further implementation in clinical practice via mobile applications.52

Ticks are today the most accepted evidence for sensitization to -Gal, but other risk factors or co-factors are likely relevant.92 Daily diet counseling, tick bite avoidance or environmental education should be firstly considered in customizing an accurate treatment for the AGS.42,61 Subsequently, an expertise behind medical interventions is required for an adequate management of the disease over time.43 Although most cases are not emergency cases, invasive techniques are required for in shock patients treatment.6 Furthermore, established protocols, symptomatology, or information about the AGS characteristics are not available due to the wide natural history and variety of subjects all over the world. Herein we present some of the most common strategies and routinary methodology in AGS treatment/management when the disease is diagnosed.

The pillar of the non-medical approach is based on avoidance. The prevention of tick bites is relevant because continuous exposure to tick bites may maintain or increase anti--Gal IgE titers and lead to allergic responses to previously tolerated foods.92 Despite limited evidence, patients who successfully avoid tick bites on a long term (12 years) have a higher chance of recovering tolerance to meat products, allowing the reintroduction of red meat into diet.10,42,93 The most common strategy for tick-bite prevention and management includes the use of lighter colored protective clothes treated with insect repellents or insecticides such as permethrin.21 Furthermore, prompt embedded tick removal using specialized fine-tipped forceps should be performed in order to reduce the amount of secreted salivary allergens.24,92

Secondly, avoidance of mammalian meat, by-products of meat (innards), fat (gelatin and lards) and other -Gal-containing foods such as dairy products represent a crucial management strategy for AGS.10,92 To achieve this goal, dietary counseling is vital, and it can be combined with nutro clinical support to avoid nutritional deficiency, especially in highly sensitized individuals.6 Patients should receive a personalized dietary follow-up depending on which foods are allergy triggering and to routinely check for iron and vitamin B12 supplementation needs.21,61

Another major foundation for AGS management is education. Vulnerable patients should be taught on nutrition facts label reading, awareness of hidden exposures and be provided with a written plan on how to promptly operate in case of an allergic reaction.6,42,61 Clinicians must also inform patients of the risk of onset anaphylaxis not only due to cetuximab but also because of heparin, gelatin-containing vaccines and mammalian heart valves.94 The fact that numerous pharmaceutical products contain animal-derived excipients makes it harder to avoid all potentially immunogenic antigens.95 For this reason, it is recommended for AGS-suffering patients to wear warning bracelets about their condition so that physicians are aware and can prevent future life-threatening situations in emergency cases.61 In fact, due to the worldwide increase in individuals with high anti--Gal IgE titers and possibly undiagnosed AGS-patients, an allergy prescreening before administration of -Gal containing medication might be recommended.96

Due to the AGS delay and unexpected symptomatology, emergency treatment is of utmost importance to correctly manage allergic reactions and potentially life-threatening anaphylaxis.6 Furthermore, the high variability regarding severity and timing of the symptoms represents a challenge for the medical management of this disease.21 Intramuscular epinephrine administration represents the initial recommendation. For patients in shock, intravenous epinephrine should be applied alongside with fluid resuscitation and occasional vasopressors. In case of airway obstruction, intubation may be necessary.97 Afterwards, in order to properly reduce the risk of a multisystem allergic reaction, it is imperative to always carry an epinephrine auto-injector.21 For tick-bite local reactions, symptomatic treatment with oral antihistamines, corticosteroids and cold compresses should be enough to reduce non-serious symptoms such as pruritus, urticaria and angioedema.98 As AGS symptomatology and severity are reported to have high individual variability and rely mostly on symptomatic treatment, information collected mostly from case reports is presented in Table 1, together with their clinical management apart from the anaphylaxis acute treatment-response already discussed. A recent study described a clinical case with abnormal neuro-psychiatric behavior (abulia, aphasia, abnormal gait, and reduction of limb movement) related to AGS and a possible -Gal driven immune-related hypothalamic dysfunction that needs further investigation.99 Other symptomatology such as palpitations and tachycardia are self-limiting and therefore resolve spontaneously.100

Table 1 Drugs and Associated Pharmacological Class Used for -Gal Syndrome Medical Treatment

As described above, humans evolved as non-capable organisms to produce the glycan -Gal.114 Together with the fact that a wide variability exists between individuals suffering from this disease,5 AGS comprehension becomes a complex goal in which molecular and physiological mechanisms need to be elucidated. Several experimental model hosts are currently available for the study of AGS and the immune response to -Gal. Zebrafish (Danio rerio) model has been established and validated under laboratory conditions. This animal model was developed by Contreras et al,115 in which allergic hemorrhagic anaphylactic-type reactions together with behavior changes and mortality were observed in response to tick salivary compounds and mammalian meat consumption. The reactions were associated with tissue-specific toll-like-receptor-mediated responses in Th1 and Th2 helper cells. These data support the use of zebrafish as an animal model for the study of the AGS and bring a new perspective for future strategies in the control of infectious diseases as reported for tuberculosis using vaccination with -Gal.116

Murine models have been used for decades as validated experimental in vivo methodology for investigating both human and animal diseases due to the advantages that these models offer in terms of time, reproducibility and genetic characteristics.117,118 However, wild-type mice produce biologically active 1,3-galactosyltransferase (1,3GT) for the synthesis of -Gal and thus lack anti-Gal antibodies.119 Knocking out the 1,3GT gene results in the absence of -Gal epitopes, not only in murine models but also in pigs,120,121 thus becoming humanized experimental animal models.

The mice C57BL/6 line is one of the most common strains used in research.122 The humanized murine model of this strain for studying AGS (GTKO, AGKO or 1,3-GalT-KO), has been used for the study of tick-induced IgE response-model for -Gal reactions,123,124 but also for testing other immunological approaches as tick-borne allergies and Chagas disease investigation.125,126

Using these animal models, further research is needed to investigate AGS risk factors and epidemiology in order to propose an accurate treatment strategy for each patient.

Mammalian meat desensitization by oral immunotherapy (OIT) has been proposed as a promising treatment for AGS as it would improve patients welfare and safer management.127 It consists of daily intake of small and generally increasing amounts of allergen, in order to reduce the immune response and consequently produce own allergen desensitization.128 To date, there are only three successful case reports (two adults and one pediatric case) of AGS with oral desensitization to beef meat.127,129 Although this type of treatment leads to a sustained unresponsiveness,93 it requires an individual effort from the patient to consume daily 120 grams of cooked mammalian meat in order to maintain desensitization,129 which also becomes an obligation and can have an impact on the patients routine, commodity and mental wellbeing. Indeed, daily mammalian meat intake could compromise the patients to develop other metabolic and cardiometabolic diseases such as hypertension, diabetes and obesity.130,131 Additionally, allergen-specific immunotherapy (AIT) using natural and recombinant -Gal containing proteins from tick sialome is also being considered for AGS treatment.17 Nonetheless, this type of therapies comes with a risk of life-threatening anaphylactic adverse reactions and demand a thoughtful and balanced management of accurate dose efficacy versus side effect appearance.66,132

Given the potential risks associated with immunotherapy, the use of allergen non-specific treatments, such as anti-IgE therapeutic monoclonal antibodies (mAbs), has found application in the treatment of food allergy.133,134 In anti-IgE therapy, mAbs bind to free serum IgE and IgE-coated B cells, acting as a competitive substrate and reducing the availability and binding between these antibodies (natural IgE) and allergy mediators such as basophils and mast cells, which increases reaction threshold and consequently reduces the risk of mild and severe anaphylactic reactions (Figure 3).43,134 Combining anti-IgE therapy as a pre-treatment with immunotherapy techniques leads to a safer administration of OIT and allows to reach the maintenance dose more rapidly.128,133,135 The anti-IgE agent omalizumab has been sporadically used in specialized centers as monotherapy in AGS patients for successfully controlling continued reactivity, allowing the introduction of a small amount of mammalian meat in their diet.42 With such a positive preliminary outcome in these patients and promising data results in other FAs (peanut and cow milk), new clinical trials using biological therapies for AGS are needed, potentially representing a future effective treatment.42,43,133

Figure 3 Anti-IgE therapy. IgE-mediated reaction with release of histamine and other co-factors occurs due to interaction of allergen-specific IgE available with IgE receptor in mediator cells (basophils, eosinophils or mast cells), which are degranulated and increase the risk of life-threatening anaphylactic adverse reactions. The pharmacological and clinical aims of the use of anti-IgEs monoclonal antibodies (mAbs) as drugs is to downregulate and/or decrease IgE production by B cells. Anti-IgE antibodies bind to both IgE-expressing B cells and free serum IgE, markedly decreasing IgE levels available for binding to IgE receptor in allergic reaction-mediator cells and, consequently, gradually compromising mast cells and basophils sensitivity to allergens.

Management of FAs is becoming less generic and more target oriented.66 Consequently, there is still a need to improve our understanding of the immunological mechanisms behind tick bite sensitization and therefore identify new and more specific targets for the development of new treatment interventions for AGS.136

Prevention from developing AGS stands on avoiding the initial -Gal sensitization caused by tick bites, being particularly beneficial for at-risk population.21,35,36 Apart from the common strategies for tick-bite prevention mentioned above, the development of tick-antigen-based vaccines could not only protect against AGS but also against other tick-borne diseases.14

Therefore, to follow the vaccinomics approach, it is essential to identify tick bioactive molecules and consequent signaling pathways that mediate tick-host-pathogen interactions.137,138 For example, in a study by Mateos-Hernndez et al,47 tick sialome proteins, with or without -Gal modifications, that led to a protective immune response and were recognized by AGS patients but not control individuals could serve as potential target antigen candidates for vaccine development. The identification of the poorly understood molecular mechanisms behind the development of spontaneous acquired tick resistance (ATR) is also of key importance as it could help in the search for new vaccine formulations.139 Discovering which tick salivary antigens are natural targets of ATR will help to aim towards the inhibition of tick feeding, reproduction and further pathogen transmission.140

AGS is an atypical, underdiagnosed vector-borne allergy that presents clinical implications beyond expected due to the presence of -Gal in various animal-derived medical products, hindering the treatment of several other pathologies.141 Since the discovery of AGS, many advancements have been made in order to obtain a better knowledge in terms of disease epidemiology, medical approach and molecular mechanisms. Nevertheless, current diagnostic methods lack specificity or are too risky for routinary appliance, creating the need to overcome these limitations with more precise methods. Also, a uniformization-based approach of diagnostic guidelines could be beneficial, creating comparable data and offering an opportunity to improve clinical decision-making accuracy. Further diagnostic, treatment and preventive advances will only be possible if the molecular and immune mechanisms behind AGS are uncovered. Furthermore, it is of utmost importance to identify tick salivary molecules, with or without -Gal modifications, that trigger IgE sensitivity as they could be the key for further vaccine development. With climate change, the tick-host paradigm will shift towards an increasing number of AGS cases in new regions worldwide,22 which will pose new challenges for clinicians in the future.

Research on AGS was funded by Ministerio de Ciencia e Innovacin/Agencia Estatal de Investigacin MCIN/AEI/10.13039/501100011033, Spain and EU-FEDER (Grant BIOGAL PID2020-116761GB-I00). R. Vaz-Rodrigues was supported by a doctoral contract (2022/20675) from Universidad de Castilla-La Mancha (UCLM), Spain, co-financed by the European Social Fund (ESF). L. Mazuecos was supported by a post-doctoral grant (2021-POST-32002) from UCLM co-financed by ESF.

The authors declare that they have no conflicts of interest in this work.

1. Hils M, Wlbing F, Hilger C, Fischer J, Hoffard N, Biedermann T. The history of carbohydrates in type I allergy. Front Immunol. 2020;11(10):114. doi:10.3389/fimmu.2020.586924

2. Hilger C, Fischer J, Wlbing F, Biedermann T. Role and mechanism of Galactose-Alpha-1,3-Galactose in the elicitation of delayed anaphylactic reactions to red meat. Curr Allergy and Asthma Rep. 2019;19(1):111. doi:10.1007/s11882-019-0835-9

3. Galili U. Evolution in primates by Catastrophic-selection interplay between enveloped virus epidemics, mutated genes of enzymes synthesizing carbohydrate antigens, and natural anti-carbohydrate antibodies. Am J Phys Anthropol. 2019;168(2):352363. doi:10.1002/ajpa.23745

4. Commins SP, Satinover SM, Hosen J, et al. Delayed anaphylaxis, angioedema, or urticaria after consumption of red meat in patients with IgE antibodies specific for galactose--1,3-galactose. J Allergy Clin Immunol. 2009;123(2):426433.e2. doi:10.1016/j.jaci.2008.10.052

5. Fischer J, Yazdi AS, Biedermann T. Clinical spectrum of -Gal syndrome: from immediate-type to delayed immediate-type reactions to mammalian innards and meat. Allergo J Int. 2016;25(2):5562. doi:10.1007/s40629-016-0099-z

6. elebiolu E, Akarsu A, ahiner M. IgE mediated food allergy throughout the life. Turk J Med Sci. 2021;51(1):4960. doi:10.3906/sag-2006-95

7. Fischer J, Reepschlger T, Schricker T, Raap U. Alpha-gal syndrome: overview of clinical presentation and pathophysiology. Hautarzt. 2022;73(3):195200. doi:10.1007/s00105-022-04943-4

8. Chinuki Y, Takahashi H, Morita E. IgE antibodies to galactose--1,3-galactose, an epitope of red meat allergen, cross-react with a novel flounder roe allergen. J Investig Allergol Clin Immunol. 2021;32(4):17. doi:10.18176/jiaci.0754

9. Dunkman WJ, Rycek W, Manning MW. What does a red meat allergy have to do with anesthesia? Perioperative management of alpha-gal syndrome. Anesth Analg. 2019;129(5):12421248. doi:10.1213/ANE.0000000000003460

10. Cabezas-Cruz A, Hodi A, Romn-Carrasco P, et al. Environmental and molecular drivers of the -Gal syndrome. Front Immunol. 2019;10(5):112. doi:10.3389/fimmu.2019.01210

11. Cabezas-Cruz A, Valds J, de la Fuente J. Cancer research meets tick vectors for infectious diseases. Lancet Infect Dis. 2014;14(10):916917. doi:10.1016/S1473-3099(14)70902-8

12. Chung CH, Mirakhur B, Chan E, et al. Cetuximab-induced anaphylaxis and IgE specific for Galactose--1,3-Galactose. N Engl J Med. 2008;358(11):11091117. doi:10.1056/nejmoa074943

13. de la Fuente J, Pacheco I, Villar M, Cabezas-Cruz A. The alpha-Gal syndrome: new insights into the tick-host conflict and cooperation. Parasit Vectors. 2019;12(1):15. doi:10.1186/s13071-019-3413-z

14. Rodrguez Y, Rojas M, Gershwin ME, Anaya JM. Tick-borne diseases and autoimmunity: a comprehensive review. J Autoimmun. 2018;88(3):2142. doi:10.1016/j.jaut.2017.11.007

15. Cabezas-Cruz A, Mateos-Hernndez L, Chmela J, Villar M, de la Fuente J. Salivary prostaglandin E2: role in tick-induced allergy to red meat. Trends Parasitol. 2017;33(7):495498. doi:10.1016/j.pt.2017.03.004

16. Romn-Carrasco P, Hemmer W, Cabezas-Cruz A, Hodi A, de la Fuente J, Swoboda I. The -Gal syndrome and potential mechanisms. Front Allergy. 2021;2(12):117. doi:10.3389/falgy.2021.783279

17. Ramasamy R. Mosquito vector proteins homologous to 1-3 galactosyl transferases of tick vectors in the context of protective immunity against malaria and hypersensitivity to vector bites. Parasit Vectors. 2021;14(1):16. doi:10.1186/s13071-021-04801-7

18. Wilson JM, Schuyler AJ, Schroeder N, Platts-Mills TAE. Galactose--1,3-Galactose: atypical food allergen or model IgE hypersensitivity? Curr Allergy Asthma Rep. 2017;17(1):39. doi:10.1007/s11882-017-0672-7

19. Cabezas-Cruz A, Hodi A, Mateos-Hernandez L, Contreras M, De La Fuente J. Tick-human interactions: from allergic klendusity to the -Gal syndrome. Biochem J. 2021;478(9):17831794. doi:10.1042/BCJ20200915

20. Carson AS, Gardner A, Iweala OI. Wheres the beef? Understanding allergic responses to red meat in alpha-gal syndrome. J Immunol. 2022;208(2):267277. doi:10.4049/jimmunol.2100712

21. Wong XL, Sebaratnam DF. Mammalian meat allergy. Int J Dermatol. 2018;57(12):14331436. doi:10.1111/ijd.14208

22. Young I, Prematunge C, Pussegoda K, Corrin T, Waddell L. Tick exposures and alpha-gal syndrome: a systematic review of the evidence. Ticks Tick Borne Dis. 2021;12(3):101674. doi:10.1016/j.ttbdis.2021.101674

23. Diaz JH. Red meat allergies after Lone Star tick (Amblyomma americanum) bites. South Med J. 2020;113(6):267274. doi:10.14423/SMJ.0000000000001102

24. van Nunen S. Tick-induced allergies: mammalian meat allergy, tick anaphylaxis and their significance. Asia Pac Allergy. 2015;5(1):316. doi:10.5415/apallergy.2015.5.1.3

25. Platts-Mills TAE, Commins SP, Biedermann T, et al. On the cause and consequences of IgE to galactose--1,3-galactose: a report from the National Institute of Allergy and Infectious Diseases Workshop on understanding IgE-mediated mammalian meat allergy. J Allergy Clin Immunol. 2020;145(4):10611071. doi:10.1016/j.jaci.2020.01.047

26. Joral A, Azketa N, Sanchez P, et al. The quantification of IgG specific to -Gal could be used as a risk marker for suffering mammalian meat allergy. Foods. 2022;11(3):466. doi:10.3390/foods11030466

27. McGain F, Welton R, Solley GO, Winkel KD. First fatalities from tick bite anaphylaxis. J Allergy Clin Immunol Pract. 2016;2(7):309314. doi:10.1080/21674086.1933.11925177

28. Mullins RJ, Wainstein BK, Barnes EH, Liew WK, Campbell DE. Increases in anaphylaxis fatalities in Australia from 1997 to 2013. Clin Exp Allergy. 2016;46(8):10991110. doi:10.1111/cea.12748

29. Commins SP. Invited commentary: alpha-gal allergy: tip of the iceberg to a pivotal immune response. Curr Allergy Asthma Rep. 2016;16(9):13. doi:10.1007/s11882-016-0641-6

30. Levin M, Apostolovic D, Biedermann T, et al. Galactose -1,3-galactose phenotypes: lessons from various patient populations. Ann Allergy Asthma Immunol. 2019;122(6):598602. doi:10.1016/j.anai.2019.03.021

31. Zurbano-Azqueta L, Antn-Casas E, Duque-Gmez S, Jimnez-Gmez I, Fernndez-Pelln L, Lpez-Gutirrez J. Alpha-gal syndrome. Allergy to red meat and gelatin. Rev Clin Esp. 2021. doi:10.1016/j.rceng.2021.06.005

32. Mateo-Borrega MB, Garcia B, Larramendi CH, et al. Ige-mediated sensitization to galactose--1,3-galactose (-gal) in urticaria and anaphylaxis in Spain: geographical variations and risk factors. J Investig Allergol Clin Immunol. 2019;29(6):436443. doi:10.18176/jiaci.0373

33. Fischer J, Lupberger E, Hebsaker J, et al. Prevalence of type I sensitization to alpha-gal in forest service employees and hunters. Allergy. 2017;72(10):15401547. doi:10.1111/all.13156

34. Villalta D, Pantarotto L, Da Re M, et al. High prevalence of sIgE to Galactose-1,3-galactose in rural pre-Alps area: a cross-sectional study. Clin Exp Allergy. 2016;46(2):377380. doi:10.1111/cea.12655

35. Mitchell CL, Lin FC, Vaughn M, Apperson CS, Meshnick SR, Commins SP. Association between Lone Star tick bites and increased alpha-gal sensitization: evidence from a prospective cohort of outdoor workers. Parasit Vectors. 2020;13(1):14. doi:10.1186/s13071-020-04343-4

36. Venturini M, Lobera T, Sebastin A, Portillo A, Oteo JA. IgE to -Gal in foresters and forest workers from La Rioja, North of Spain. J Investig Allergol Clin Immunol. 2018;28(2):106112. doi:10.18176/jiaci.0218

37. Cabezas-Cruz A, de la Fuente J, Fischer J. Prevalence of type I sensitization to alpha-gal in forest service employees and hunters: is the blood type an overlooked risk factor in epidemiological studies of the -Gal syndrome? Allergy. 2017;72(12):20442047. doi:10.1111/all.13206

38. Cabezas-Cruz A, Mateos-Hernndez L, Alberdi P, et al. Effect of blood type on anti-a-Gal immunity and the incidence of infectious diseases. Exp Mol Med. 2017;49(3):e301e301. doi:10.1038/emm.2016.164

39. Yilmaz B, Portugal S, Tran TM, et al. Gut microbiota elicits a protective immune response against malaria transmission. Cell. 2014;159(6):12771289. doi:10.1016/j.cell.2014.10.053

40. Urra JM, Ferreras-Colino E, Contreras M, et al. The antibody response to the glycan -Gal correlates with COVID-19 disease symptoms. J Med Virol. 2021;93(4):20652075. doi:10.1002/jmv.26575

41. Steinke JW, Platts-Mills TAE, Commins SP. The alpha-gal story: lessons learned from connecting the dots. J Allergy Clin Immunol. 2015;135(3):589596. doi:10.1016/j.jaci.2014.12.1947

42. Commins SP. Diagnosis & management of alpha-gal syndrome: lessons from 2500 patients. Expert Rev Clin Immunol. 2020;16(7):667677. doi:10.1080/1744666X.2020.1782745

43. Lopes JP, Sicherer S. Food allergy: epidemiology, pathogenesis, diagnosis, prevention, and treatment. Curr Opin Immunol. 2020;66:5764. doi:10.1016/j.coi.2020.03.014

44. Antunes J, Borrego L, Romeira A, Pinto P. Skin prick tests and allergy diagnosis. Allergol Immunopathol. 2009;37(3):155164. doi:10.1016/S0301-0546(09)71728-8

45. Shroba J, Rath N, Barnes C. Possible role of environmental factors in the development of food allergies. Clin Rev Allergy Immunol. 2019;57(3):303311. doi:10.1007/s12016-018-8703-2

46. Jacquenet S, Moneret-Vautrin DA, Bihain BE. Mammalian meat-induced anaphylaxis: clinical relevance of anti-galactose--1,3-galactose IgE confirmed by means of skin tests to cetuximab. J Allergy Clin Immunol. 2009;124(3):603605. doi:10.1016/j.jaci.2009.06.014

47. Mateos-Hernndez L, Villar M, Moral A, et al. Tick-host conflict: immunoglobulin E antibodies to tick proteins in patients with anaphylaxis to tick bite. Oncotarget. 2017;8(13):2063020644. doi:10.18632/oncotarget.15243

48. Hocaoglu AB, Cipe F, Aydogmus C. Are skin prick tests really safe? A case of anaphylaxis caused by skin prick testing with inhalant allergens. Allergol Immunopathol. 2015;43(2):215216. doi:10.1016/j.aller.2013.09.011

49. Alns M. Anaphylaxis following prick-by-prick testing with peanut. Clin Case Rep. 2020;8(12):23662368. doi:10.1002/ccr3.3154

50. Chiriac AM, Bousquet J, Demoly P. In vivo methods for the study and diagnosis of allergy. In: Burks AW, Holgate ST, OHehir RE, et al. editors. Middletons Allergy: Principles and Practice. 9th ed. Elsevier; 2020:11191132. doi:10.1016/B978-0-323-08593-9.00071-1

51. Calvani M, Bianchi A, Reginelli C, Peresso M, Testa A. Oral food challenge. Medicina. 2019;55(10):116. doi:10.3390/medicina55100651

52. de la Fuente J, Cabezas-Cruz A, Pacheco I. Alpha-gal syndrome: challenges to understanding sensitization and clinical reactions to alpha-gal. Expert Rev Mol Diagn. 2020;20(9):905911. doi:10.1080/14737159.2020.1792781

53. Mehlich J, Fischer J, Hilger C, et al. The basophil activation test differentiates between patients with alpha-gal syndrome and asymptomatic alpha-gal sensitization. J Allergy Clin Immunol. 2019;143(1):182189. doi:10.1016/j.jaci.2018.06.049

54. Mabelane T, Basera W, Botha M, Thomas HF, Ramjith J, Levin ME. Predictive values of alpha-gal IgE levels and alpha-gal IgE: total IgE ratio and oral food challenge proven meat allergy in a population with a high prevalence of reported red meat allergy. Pediatr Allergy Immunol. 2018;29(8):841849. doi:10.1111/pai.12969

55. Eberlein B, Mehlich J, Reidenbach K, et al. Negative oral provocation test with porcine pancreatic enzyme plus cofactors despite confirmed -gal syndrome. J Investig Allergol Clin Immunol. 2020;30(6):468469. doi:10.18176/jiaci.0513

56. Morisset M, Richard C, Astier C, et al. Anaphylaxis to pork kidney is related to IgE antibodies specific for galactose-alpha-1,3-galactose. Allergy. 2012;67(5):699704. doi:10.1111/j.1398-9995.2012.02799.x

57. Fischer J, Biedermann T. Delayed immediate-type hypersensitivity to red meat and innards: current insights into a novel disease entity. J Dtsch Dermatol Ges. 2016;14(1):3843. doi:10.1111/ddg.12821

58. Jappe U, Minge S, Kreft B, et al. Meat allergy associated with galactosyl--(1,3)-galactose (-Gal)-Closing diagnostic gaps by anti--Gal IgE immune profiling. Allergy. 2018;73(1):93105. doi:10.1111/all.13238

59. Leung DYM, Szefler SJ. Tick bites: a common cause of IgE antibodies to alpha-gal. J Allergy Clin Immunol. 2011;127(5):11311132. doi:10.1016/j.jaci.2011.03.019

60. Chinuki Y, Morita E. Alpha-Gal-containing biologics and anaphylaxis. Allergol Int. 2019;68(3):296300. doi:10.1016/j.alit.2019.04.001

61. Mabelane T, Ogunbanjo GA. Ingestion of mammalian meat and alpha-gal allergy: clinical relevance in primary care. Afr J Prim Health Care Fam Med. 2019;11(1):15. doi:10.4102/phcfm.v11i1.1901

62. Park Y, Kim D, Boorgula GD, et al. Alpha-gal and cross-reactive carbohydrate determinants in the N-Glycans of salivary glands in the Lone Star Tick, Amblyomma americanum. Vaccines. 2020;8(1):116. doi:10.3390/vaccines8010018

63. Apostolovic D, Grundstrm J, Perusko M, et al. Course of IgE to -Gal in a Swedish population of -Gal syndrome patients. Clin Transl Allergy. 2021;11(10):24. doi:10.1002/clt2.12087

64. Gonzalez-Quintela A, Dam Laursen AS, Vidal C, Skaaby T, Gude F, Linneberg A. IgE antibodies to alpha-gal in the general adult population: relationship with tick bites, atopy, and cat ownership. Clin Exp Allergy. 2014;44(8):10611068. doi:10.1111/cea.12326

65. Wilson JM, Keshavarz B, Retterer M, et al. A dynamic relationship between two regional causes of IgE-mediated anaphylaxis: -Gal syndrome and imported fire ant. J Allergy Clin Immunol. 2021;147(2):643652.e7. doi:10.1016/j.jaci.2020.05.034

66. Passanisi S, Lombardo F, Crisafulli G, Salzano G, Aversa T, Pajno GB. Novel diagnostic techniques and therapeutic strategies for IgE-mediated food allergy. Allergy Asthma Proc. 2021;42(2):124130. doi:10.2500/AAP.2021.42.200129

Read this article:
Diagnosis and treatment of the alpha-Gal syndrome | JAA - Dove Medical Press

Posted in Integrative Medicine | Comments Off on Diagnosis and treatment of the alpha-Gal syndrome | JAA – Dove Medical Press

Umoja Biopharma and TreeFrog Therapeutics Announce Collaboration to Address Current Challenges Facing Ex Vivo Allogeneic Therapies in Immuno-Oncology…

Posted: June 13, 2022 at 2:06 am

SEATTLE and PESSAC, France, June 10, 2022 (GLOBE NEWSWIRE) -- Umoja Biopharma, Inc., an immuno-oncology company pioneering off-the-shelf, integrated therapeutics that reprogram immune cells to treat patients with solid and hematologic malignancies, and TreeFrog Therapeutics, a biotechnology company aimed at making safer, more efficient and more affordable cell therapies based on induced pluripotent stem cells (iPSCs), announced today that they have entered into a collaboration to evaluate Umojas iPSC platform within TreeFrogs C-Stem technology for scalable expansion and immune cell differentiation in bioreactors.

Together, the successful pairing of Umojas RACR engineered iPS cells and TreeFrogs C-Stem technology could overcome several challenges facing ex vivo allogeneic therapies, said Ryan Larson, Ph.D., Vice President and Head of Translational Science at Umoja. Two major industry-wide challenges include the ability to scale iPSC-based culture while maintaining cell health, quality, and efficient immune cell differentiation. TreeFrogs biomimetic C-Stem technology is the perfect complementary development platform for our RACR technology, a pairing which could result in controlled, efficient iPSC expansion and differentiation into immune cells, with improved yields and quality. In addition to enhancing the differentiation and yield of immune cells within the manufacturing process, our RACR system should bring therapeutic benefit to patients, allowing for safe in vivo engraftment and persistence of tumor-killing cells without requirements for toxic lymphodepleting chemotherapy.

Umoja is developing an engineered iPSC platform that addressesmany challenges associated with ex vivo cell therapy manufacturing, including limited scalability and manufacturing complexity.Umojas iPSCs are engineered with a synthetic rapamycin-activated cytokine receptor (RACR) to drive differentiation to, and expansion of innate cytotoxic lymphoid cells, including but not limited to natural killer (NK) cells in the absence of exogenous cytokines and feeder cells. TreeFrogs proprietary C-Stem technology relies on the high-throughput encapsulation (>1,000 capsules/second) of hiPSCs within biomimetic alginate shells, which promote in vivo-like exponential growth and protect cells from external stress. In 2021, C-Stem was demonstrated to allow for unprecedented iPSC expansion in 10L bioreactors, while preserving stem cell quality. Also enabling direct in-capsule iPSC differentiation, C-Stem constitutes a scalable, end-to-end, and GMP-compatible manufacturing platform for iPSC-derived cell therapies.

Frdric Desdouits, Ph.D., Chief Executive Officer at TreeFrog added, Our primary goal is to bring the benefits of the C-Stem technology to patients as fast as possible, either through in-house programs or strategic alliances with cell therapy leaders. Partnering with Umoja is an important step forward in immuno-oncology. Besides scale-up and cell quality, the in vivo persistence of allogeneic therapies remains a critical challenge in the industry. We believe Umojas platform will allow for safer and more efficient allogeneic cell therapies in immuno-oncology. We look forward to rapidly advancing this joint approach to clinic and contributing to the future of off-the-shelf cancer treatments.

About Umoja BiopharmaUmoja Biopharma, Inc. is an early clinical-stage company advancing an entirely new approach to immunotherapy. Umoja Biopharma, Inc. is a transformative multi-platform immuno-oncology company founded with the goal of creating curative treatments for solid and hematological malignancies by reprogramming immune cells in vivo to target and fight cancer. Founded based on pioneering work performed at Seattle Childrens Research Institute and Purdue University, Umojas novel approach is powered by integrated cellular immunotherapy technologies including the VivoVec in vivo delivery platform, the RACR/CAR in vivo cell expansion/control platform, and the TumorTag targeting platform. Designed from the ground up to work together, these platforms are being developed to create and harness a powerful immune response in the body to directly, safely, and controllably attack cancer. Umoja believes that its approach can provide broader access to the most advanced immunotherapies and enable more patients to live better, fuller lives. To learn more, visithttp://umoja-biopharma.com/.

About TreeFrog TherapeuticsTreeFrog Therapeutics is a French-based biotech company aiming to unlock access to cell therapies for millions of patients. TreeFrog Therapeutics is developing a pipeline of therapeutic candidates using proprietary C-Stem technology, allowing for the mass production of induced pluripotent stem cells and their differentiation into ready-to-transplant microtissues with unprecedented scalability and cell quality. Bringing together over 80 biophysicists, cell biologists and bioproduction engineers, TreeFrog Therapeutics raised $82M over the past 3 years to advance its pipeline in regenerative medicine and immuno-oncology. The company is currently opening technological hubs in Boston, USA, and Kobe, Japan, to drive the adoption of C-Stem and build strategic alliances with leading academic, biotech and industry players in the field of cell therapy.

Umoja Biopharma Media Contact:Darren Opland, Ph.D.LifeSci Communicationsdarren@lifescicomms.com

TreeFrog Therapeutics Media Contact:Pierre-Emmanuel GaultierTreeFrog Therapeuticspierre@treefrog.fr

A photo accompanying this announcement is available at https://www.globenewswire.com/NewsRoom/AttachmentNg/012ae87d-b7c6-4fa2-81dc-c769877b182c

Link:
Umoja Biopharma and TreeFrog Therapeutics Announce Collaboration to Address Current Challenges Facing Ex Vivo Allogeneic Therapies in Immuno-Oncology...

Posted in Cell Therapy | Comments Off on Umoja Biopharma and TreeFrog Therapeutics Announce Collaboration to Address Current Challenges Facing Ex Vivo Allogeneic Therapies in Immuno-Oncology…

Frontiers | Nanomedicine: Principles, Properties, and …

Posted: January 5, 2022 at 2:41 am

Introduction

Over the last years, nanotechnology has been introduced in our daily routine. This revolutionary technology has been applied in multiple fields through an integrated approach. An increasing number of applications and products containing nanomaterials or at least with nano-based claims have become available. This also happens in pharmaceutical research. The use of nanotechnology in the development of new medicines is now part of our research and in the European Union (EU) it has been recognized as a Key Enabling Technology, capable of providing new and innovative medical solution to address unmet medical needs (Bleeker et al., 2013; Ossa, 2014; Tinkle et al., 2014; Pita et al., 2016).

The application of nanotechnology for medical purposes has been termed nanomedicine and is defined as the use of nanomaterials for diagnosis, monitoring, control, prevention and treatment of diseases (Tinkle et al., 2014). However, the definition of nanomaterial has been controversial among the various scientific and international regulatory corporations. Some efforts have been made in order to find a consensual definition due to the fact that nanomaterials possess novel physicochemical properties, different from those of their conventional bulk chemical equivalents, due to their small size. These properties greatly increase a set of opportunities in the drug development; however, some concerns about safety issues have emerged. The physicochemical properties of the nanoformulation which can lead to the alteration of the pharmacokinetics, namely the absorption, distribution, elimination, and metabolism, the potential for more easily cross biological barriers, toxic properties and their persistence in the environment and human body are some examples of the concerns over the application of the nanomaterials (Bleeker et al., 2013; Tinkle et al., 2014).

To avoid any concern, it is necessary establishing an unambiguous definition to identify the presence of nanomaterials. The European Commission (EC) created a definition based on the European Commission Joint Research Center and on the Scientific Committee on Emerging and Newly Identified Health Risks. This definition is only used as a reference to determine whether a material is considered a nanomaterial or not; however, it is not classified as hazardous or safe. The EC claims that it should be used as a reference for additional regulatory and policy frameworks related to quality, safety, efficacy, and risks assessment (Bleeker et al., 2013; Boverhof et al., 2015).

According to the EC recommendation, nanomaterial refers to a natural, incidental, or manufactured material comprising particles, either in an unbound state or as an aggregate wherein one or more external dimensions is in the size range of 1100 nm for 50% of the particles, according to the number size distribution. In cases of environment, health, safety or competitiveness concern, the number size distribution threshold of 50% may be substituted by a threshold between 1 and 50%. Structures with one or more external dimensions below 1 nm, such as fullerenes, graphene flakes, and single wall carbon nanotubes, should be considered as nanomaterials. Materials with surface area by volume in excess of 60 m2/cm3 are also included (Commission Recommendation., 2011). This defines a nanomaterial in terms of legislation and policy in the European Union. Based on this definition, the regulatory bodies have released their own guidances to support drug product development.

The EMA working group introduces nanomedicines as purposely designed systems for clinical applications, with at least one component at the nanoscale, resulting in reproducible properties and characteristics, related to the specific nanotechnology application and characteristics for the intended use (route of administration, dose), associated with the expected clinical advantages of nano-engineering (e.g., preferential organ/tissue distribution; Ossa, 2014).

Food and Drug Administration (FDA) has not established its own definition for nanotechnology, nanomaterial, nanoscale, or other related terms, instead adopting the meanings commonly employed in relation to the engineering of materials that have at least one dimension in the size range of approximately 1 nanometer (nm) to 100 nm. Based on the current scientific and technical understanding of nanomaterials and their characteristics, FDA advises that evaluations of safety, effectiveness, public health impact, or regulatory status of nanotechnology products should consider any unique properties and behaviors that the application of nanotechnology may impart (Guidance for Industry, FDA, 2014).

According to the former definition, there are three fundamental aspects to identify the presence of a nanomaterial, which are size, particle size distribution (PSD) and surface area (Commission Recommendation., 2011; Bleeker et al., 2013; Boverhof et al., 2015).

The most important feature to take into account is size, because it is applicable to a huge range of materials. The conventional range is from 1 to 100 nm. However, there is no bright line to set this limit. The maximum size that a material can have to be considered nanomaterial is an arbitrary value because the psychochemical and biological characteristics of the materials do not change abruptly at 100 nm. To this extent, it is assumed that other properties should be taken in account (Lvestam et al., 2010; Commission Recommendation., 2011; Bleeker et al., 2013; Boverhof et al., 2015).

The pharmaceutical manufacturing of nanomaterials involves two different approaches: top down and bottom down. The top down process involves the breakdown of a bulk material into a smaller one or smaller pieces by mechanical or chemical energy. Conversely, the bottom down process starts with atomic or molecular species allowing the precursor particles to increase in size through chemical reaction (Luther, 2004; Oberdrster, 2010; Boverhof et al., 2015). These two processes of manufacturing are in the origin of different forms of particles termed primary particle, aggregate and agglomerate (Figure 1). The respective definition is (sic):

Figure 1. Schematic representation of the different forms of particles: primary particle, aggregate, and agglomerate (reproduced with permission from Oberdrster, 2010).

particle is a minute piece of matter with defined physical boundaries (Oberdrster, 2010; Commission Recommendation., 2011);

aggregate denotes a particle comprising strongly bound or fused particlesand the external surface can be smaller than the sum of the surface areas of the individual particles (Oberdrster, 2010; Commission Recommendation., 2011);

agglomerate means a collection of weakly bound particles or aggregates where the resulting external surface area are similar to the sum of the surface areas of the individual components (Oberdrster, 2010; Commission Recommendation., 2011).

Considering the definition, it is understandable why aggregates and agglomerates are included. They may still preserve the properties of the unbound particles and have the potential to break down in to nanoscale (Lvestam et al., 2010; Boverhof et al., 2015). The lower size limit is used to distinguish atoms and molecules from particles (Lvestam et al., 2010).

The PSD is a parameter widely used in the nanomaterial identification, reflecting the range of variation of sizes. It is important to set the PSD, because a nanomaterial is usually polydisperse, which means, it is commonly composed by particles with different sizes (Commission Recommendation., 2011; Bleeker et al., 2013; Boverhof et al., 2015).

The determination of the surface area by volume is a relational parameter, which is necessary when requested by additional legislation. The material is under the definition if the surface area by volume is larger than 60 m2/cm3, as pointed out. However, the PSD shall prevail, and for example, a material is classified as a nanomaterial based on the particle size distribution, even if the surface area by volume is lower than the specified 60 m2/cm3 (Commission Recommendation., 2011; Bleeker et al., 2013; Boverhof et al., 2015).

Nanomaterials can be applied in nanomedicine for medical purposes in three different areas: diagnosis (nanodiagnosis), controlled drug delivery (nanotherapy), and regenerative medicine. A new area which combines diagnostics and therapy termed theranostics is emerging and is a promising approach which holds in the same system both the diagnosis/imaging agent and the medicine. Nanomedicine is holding promising changes in clinical practice by the introduction of novel medicines for both diagnosis and treatment, having enabled to address unmet medical needs, by (i) integrating effective molecules that otherwise could not be used because of their high toxicity (e.g., Mepact), (ii) exploiting multiple mechanisms of action (e.g., Nanomag, multifunctional gels), (iii) maximizing efficacy (e.g., by increasing bioavailability) and reducing dose and toxicity, (iv) providing drug targeting, controlled and site specific release, favoring a preferential distribution within the body (e.g., in areas with cancer lesions) and improved transport across biological barriers (Chan, 2006; Mndez-Rojas et al., 2009; Zhang et al., 2012; Ossa, 2014).

This is a result of intrinsic properties of nanomaterials that have brought many advantages in the pharmaceutical development. Due to their small size, nanomaterials have a high specific surface area in relation to the volume. Consequently, the particle surface energy is increased, making the nanomaterials much more reactive. Nanomaterials have a tendency to adsorb biomolecules, e.g., proteins, lipids, among others, when in contact with the biological fluids. One of the most important interactions with the living matter relies on the plasma/serum biomoleculeadsorption layer, known as corona, that forms on the surface of colloidal nanoparticles (Pino et al., 2014). Its composition is dependent on the portal of entry into the body and on the particular fluid that the nanoparticles come across with (e.g., blood, lung fluid, gastro-intestinal fluid, etc.). Additional dynamic changes can influence the corona constitution as the nanoparticle crosses from one biological compartment to another one (Pearson et al., 2014; Louro, 2018).

Furthermore, optical, electrical and magnetic properties can change and be tunable through electron confinement in nanomaterials. In addition, nanomaterials can be engineered to have different size, shape, chemical composition and surface, making them able to interact with specific biological targets (Oberdrster et al., 2005; Kim et al., 2010). A successful biological outcome can only be obtained resorting to careful particle design. As such, a comprehensive knowledge of how the nanomaterials interact with biological systems are required for two main reasons.

The first one is related to the physiopathological nature of the diseases. The biological processes behind diseases occur at the nanoscale and can rely, for example, on mutated genes, misfolded proteins, infection by virus or bacteria. A better understanding of the molecular processes will provide the rational design on engineered nanomaterials to target the specific site of action desired in the body (Kim et al., 2010; Albanese et al., 2012). The other concern is the interaction between nanomaterial surface and the environment in biological fluids. In this context, characterization of the biomolecules corona is of utmost importance for understanding the mutual interaction nanoparticle-cell affects the biological responses. This interface comprises dynamic mechanisms involving the exchange between nanomaterial surfaces and the surfaces of biological components (proteins, membranes, phospholipids, vesicles, and organelles). This interaction stems from the composition of the nanomaterial and the suspending media. Size, shape, surface area, surface charge and chemistry, energy, roughness, porosity, valence and conductance states, the presence of ligands, or the hydrophobic/ hydrophilic character are some of the material characteristics that influence the respective surface properties. In turn, the presence of water molecules, acids and bases, salts and multivalent ions, surfactants are some of the factors related to the medium that will influence the interaction. All these aspects will govern the characteristics of the interface between the nanomaterial and biological components and, consequently, promote different cellular fates (Nel et al., 2009; Kim et al., 2010; Albanese et al., 2012; Monopoli et al., 2012).

A deeper knowledge about how the physicochemical properties of the biointerface influence the cellular signaling pathway, kinetics and transport will thus provide critical rules to the design of nanomaterials (Nel et al., 2009; Kim et al., 2010; Albanese et al., 2012; Monopoli et al., 2012).

The translation of nanotechnology form the bench to the market imposed several challenges. General issues to consider during the development of nanomedicine products including physicochemical characterization, biocompatibility, and nanotoxicology evaluation, pharmacokinetics and pharmacodynamics assessment, process control, and scale-reproducibility (Figure 2) are discussed in the sections that follow.

Figure 2. Schematic representation of the several barriers found throughout the development of a nanomedicine product.

The characterization of a nanomedicine is necessary to understand its behavior in the human body, and to provide guidance for the process control and safety assessment. This characterization is not consensual in the number of parameters required for a correct and complete characterization. Internationally standardized methodologies and the use of reference nanomaterials are the key to harmonize all the different opinions about this topic (Lin et al., 2014; Zhao and Chen, 2016).

Ideally, the characterization of a nanomaterial should be carried out at different stages throughout its life cycle, from the design to the evaluation of its in vitro and in vivo performance. The interaction with the biological system or even the sample preparation or extraction procedures may modify some properties and interfere with some measurements. In addition, the determination of the in vivo and in vitro physicochemical properties is important for the understanding of the potential risk of nanomaterials (Lin et al., 2014; Zhao and Chen, 2016).

The Organization for Economic Co-operation and Development started a Working Party on Manufactured Nanomaterials with the International Organization for Standardization to provide scientific advice for the safety use of nanomaterials that include the respective physicochemical characterization and the metrology. However, there is not an effective list of minimum parameters. The following characteristics should be a starting point to the characterization: particle size, shape and size distribution, aggregation and agglomeration state, crystal structure, specific surface area, porosity, chemical composition, surface chemistry, charge, photocatalytic activity, zeta potential, water solubility, dissolution rate/kinetics, and dustiness (McCall et al., 2013; Lin et al., 2014).

Concerning the chemical composition, nanomaterials can be classified as organic, inorganic, crystalline or amorphous particles and can be organized as single particles, aggregates, agglomerate powders or dispersed in a matrix which give rise to suspensions, emulsions, nanolayers, or films (Luther, 2004).

Regarding dimension, if a nanomaterial has three dimensions below 100 nm, it can be for example a particle, a quantum dot or hollow sphere. If it has two dimensions below 100 nm it can be a tube, fiber or wire and if it has one dimension below 100 nm it can be a film, a coating or a multilayer (Luther, 2004).

Different techniques are available for the analysis of these parameters. They can be grouped in different categories, involving counting, ensemble, separation and integral methods, among others (Linsinger et al., 2012; Contado, 2015).

Counting methods make possible the individualization of the different particles that compose a nanomaterial, the measurement of their different sizes and visualization of their morphology. The particles visualization is preferentially performed using microscopy methods, which include several variations of these techniques. Transmission Electron Microscopy (TEM), High-Resolution TEM, Scanning Electron Microscopy (SEM), cryo-SEM, Atomic Force Microscopy and Particle Tracking Analysis are just some of the examples. The main disadvantage of these methods is the operation under high-vacuum, although recently with the development of cryo-SEM sample dehydration has been prevented under high-vacuum conditions (Linsinger et al., 2012; Contado, 2015; Hodoroaba and Mielke, 2015).

These methods involve two steps of sample treatment: the separation of the particles into a monodisperse fraction, followed by the detection of each fraction. Field-Flow Fractionation (FFF), Analytical Centrifugation (AC) and Differential Electrical Mobility Analysis are some of the techniques that can be applied. The FFF techniques include different methods which separate the particles according to the force field applied. AC separates the particles through centrifugal sedimentation (Linsinger et al., 2012; Contado, 2015; Hodoroaba and Mielke, 2015).

Ensemble methods allow the report of intensity-weighted particle sizes. The variation of the measured signal over time give the size distribution of the particles extracted from a combined signal. Dynamic Light Scattering (DLS), Small-angle X-ray Scattering (SAXS) and X-ray Diffraction (XRD) are some of the examples. DLS and QELS are based on the Brownian motion of the sample. XRD is a good technique to obtain information about the chemical composition, crystal structure and physical properties (Linsinger et al., 2012; Contado, 2015; Hodoroaba and Mielke, 2015).

The integral methods only measure an integral property of the particle and they are mostly used to determine the specific surface area. Brunauer Emmet Teller is the principal method used and is based on the adsorption of an inert gas on the surface of the nanomaterial (Linsinger et al., 2012; Contado, 2015; Hodoroaba and Mielke, 2015).

Other relevant technique is the electrophoretic light scattering (ELS) used to determine zeta potential, which is a parameter related to the overall charge a particle acquires in a particular medium. ELS measures the electrophoretic mobility of particles in dispersion, based on the principle of electrophoresis (Linsinger et al., 2012).

The Table 1 shows some of principal methods for the characterization of the nanomaterials including the operational principle, physicochemical parameters analyzed and respective limitations.

Another challenge in the pharmaceutical development is the control of the manufacturing process by the identification of the critical parameters and technologies required to analyse them (Gaspar, 2010; Gaspar et al., 2014; Sainz et al., 2015).

New approaches have arisen from the pharmaceutical innovation and the concern about the quality and safety of new medicines by regulatory agencies (Gaspar, 2010; Gaspar et al., 2014; Sainz et al., 2015).

Quality-by-Design (QbD), supported by Process Analytical Technologies (PAT) is one of the pharmaceutical development approaches that were recognized for the systematic evaluation and control of nanomedicines (FDA, 2004; Gaspar, 2010; Gaspar et al., 2014; Sainz et al., 2015; European Medicines Agency, 2017).

Note that some of the physicochemical characteristics of nanomaterials can change during the manufacturing process, which compromises the quality and safety of the final nanomedicine. The basis of QbD relies on the identification of the Quality Attributes (QA), which refers to the chemical, physical or biological properties or another relevant characteristic of the nanomaterial. Some of them may be modified by the manufacturing and should be within a specific range for quality control purposes. In this situation, these characteristics are considered Critical Quality Attributes (CQA). The variability of the CQA can be caused by the critical material attributes and process parameters (Verma et al., 2009; Riley and Li, 2011; Bastogne, 2017; European Medicines Agency, 2017).

The quality should not be tested in nanomedicine, but built on it instead, by the understanding of the therapeutic purpose, pharmacological, pharmacokinetic, toxicological, chemical and physical properties of the medicine, process formulation, packaging, and the design of the manufacturing process. This new approach allows better focus on the relevant relationships between the characteristics, parameters of the formulation and process in order to develop effective processes to ensure the quality of the nanomedicines (FDA, 2014).

According to the FDA definition PAT is a system for designing, analzsing, and controlling manufacturing through timely measurements (i.e., during processing) of critical quality and performance attributes of raw and in-process materials and processes, with the goal of ensuring final product quality (FDA, 2014). The PAT tools analyse the critical quality and performance attributes. The main point of the PAT is to assure and enhance the understanding of the manufacturing concept (Verma et al., 2009; Riley and Li, 2011; FDA, 2014; Bastogne, 2017; European Medicines Agency, 2017).

Biocompatibility is another essential property in the design of drug delivery systems. One very general and brief definition of a biocompatible surface is that it cannot trigger an undesired' response from the organism. Biocompatibility is alternatively defined as the ability of a material to perform with an appropriate response in a specific application (Williams, 2003; Keck and Mller, 2013).

Pre-clinical assessment of nanomaterials involve a thorough biocompatibility testing program, which typically comprises in vivo studies complemented by selected in vitro assays to prove safety. If the biocompatibility of nanomaterials cannot be warranted, potentially advantageous properties of nanosystems may raise toxicological concerns.

Regulatory agencies, pharmaceutical industry, government, and academia are making efforts to accomplish specific and appropriate guidelines for risk assessment of nanomaterials (Hussain et al., 2015).

In spite of efforts to harmonize the procedures for safety evaluation, nanoscale materials are still mostly treated as conventional chemicals, thus lacking clear specific guidelines for establishing regulations and appropriate standard protocols. However, several initiatives, including scientific opinions, guidelines and specific European regulations and OECD guidelines such as those for cosmetics, food contact materials, medical devices, FDA regulations, as well as European Commission scientific projects (NanoTEST project, http://www.nanotest-fp7.eu) specifically address nanomaterials safety (Juillerat-Jeanneret et al., 2015).

In this context, it is important to identify the properties, to understand the mechanisms by which nanomaterials interact with living systems and thus to understand exposure, hazards and their possible risks.

Note that the pharmacokinetics and distribution of nanoparticles in the body depends on their surface physicochemical characteristics, shape and size. For example, nanoparticles with 10 nm in size were preferentially found in blood, liver, spleen, kidney, testis, thymus, heart, lung, and brain, while larger particles are detected only in spleen, liver, and blood (De Jong et al., 2008; Adabi et al., 2017).

In turn, the surface of nanoparticles also impacts upon their distribution in these organs, since their combination with serum proteins available in systemic circulation, influencing their cellular uptake. It should be recalled that a biocompatible material generates no immune response. One of the cause for an immune response can rely on the adsorption pattern of body proteins. An assessment of the in vivo protein profile is therefore crucial to address these interactions and to establish biocompatibility (Keck et al., 2013).

Finally, the clearance of nanoparticles is also size and surface dependent. Small nanoparticles, bellow 2030 nm, are rapidly cleared by renal excretion, while 200 nm or larger particles are more efficiently taken up by mononuclear phagocytic system (reticuloendothelial system) located in the liver, spleen, and bone marrow (Moghimi et al., 2001; Adabi et al., 2017).

Studies are required to address how nanomaterials penetrate cells and tissues, and the respective biodistribution, degradation, and excretion.

Due to all these issues, a new field in toxicology termed nanotoxicology has emerged, which aims at studying the nanomaterial effects deriving from their interaction with biological systems (Donaldson et al., 2004; Oberdrster, 2010; Fadeel, 2013).

The evaluation of possible toxic effects of the nanomaterials can be ascribed to the presence of well-known molecular responses in the cell. Nanomaterials are able to disrupt the balance of the redox systems and, consequently, lead to the production of reactive species of oxygen (ROS). ROS comprise hydroxyl radicals, superoxide anion and hydrogen peroxide. Under normal conditions, the cells produce these reactive species as a result of the metabolism. However, when exposed to nanomaterials the production of ROS increases. Cells have the capacity to defend itself through reduced glutathione, superoxide dismutase, glutathione peroxidase and catalase mechanisms. The superoxide dismutase converts superoxide anion into hydrogen peroxide and catalase, in contrast, converts it into water and molecular oxygen (Nel et al., 2006; Arora et al., 2012; Azhdarzadeh et al., 2015). Glutathione peroxidase uses glutathione to reduce some of the hydroperoxides. Under normal conditions, the glutathione is almost totally reduced. Nevertheless, an increase in ROS lead to the depletion of the glutathione and the capacity to neutralize the free radicals is decreased. The free radicals will induce oxidative stress and interact with the fatty acids in the membranes of the cell (Nel et al., 2006; Arora et al., 2012; Azhdarzadeh et al., 2015).

Consequently, the viability of the cell will be compromised by the disruption of cell membranes, inflammation responses caused by the upregulation of transcription factors like the nuclear factor kappa , activator protein, extracellular signal regulated kinases c-Jun, N-terminal kinases and others. All these biological responses can result on cell apoptosis or necrosis. Distinct physiological outcomes are possible due to the different pathways for cell injury after the interaction between nanomaterials and cells and tissues (Nel et al., 2006; Arora et al., 2012; Azhdarzadeh et al., 2015).

Over the last years, the number of scientific publications regarding toxicological effects of nanomaterials have increased exponentially. However, there is a big concern about the results of the experiments, because they were not performed following standard and harmonized protocols. The nanomaterial characterization can be considered weak once there are not standard nanomaterials to use as reference and the doses used in the experiences sometimes cannot be applied in the biological system. Therefore, the results are not comparable. For a correct comparison, it is necessary to perform a precise and thorough physicochemical characterization to define risk assessment guidelines. This is the first step for the comparison between data from biological and toxicological experiments (Warheit, 2008; Fadeel et al., 2015; Costa and Fadeel, 2016).

Although nanomaterials may have an identical composition, slight differences e.g., in the surface charge, size, or shape could impact on their respective activity and, consequently, on their cellular fate and accumulation in the human body, leading to different biological responses (Sayes and Warheit, 2009).

Sayes and Warheit (2009) proposed a three phases model for a comprehensive characterization of nanomaterials. Accordingly, the primary phase is achieved in the native state of the nanomaterial, specifically, in its dry state. The secondary characterization is performed with the nanomaterials in the wet phase, e.g., as solution or suspension. The tertiary characterization includes in vitro and in vivo interactions with biological systems. The tertiary characterization is the most difficult from the technical point of view, especially in vivo, because of all the ethical questions concerning the use of animals in experiments (Sayes and Warheit, 2009).

Traditional toxicology uses of animals to conduct tests. These types of experiments using nanomaterials can be considered impracticable and unethical. In addition, it is time-consuming, expensive and sometimes the end points achieved are not enough to correctly correlate with what happens in the biological systems of animals and the translation to the human body (Collins et al., 2017).

In vitro studies are the first assays used for the evaluation of cytotoxicity. This approach usually uses cell lines, primary cells from the tissues, and/or a mixture of different cells in a culture to assess the toxicity of the nanomaterials. Different in vitro cytotoxicity assays to the analysis of the cell viability, stress, and inflammatory responses are available. There are several cellular processes to determine the cell viability, which consequently results in different assays with distinct endpoints. The evaluation of mitochondrial activity, the lactate dehydrogenase release from the cytosol by tretazolium salts and the detection of the biological marker Caspase-3 are some of the examples that imposes experimental variability in this analysis. The stress response is another example which can be analyzed by probes in the evaluation of the inflammatory response via enzyme linked immunosorbent assay are used (Kroll et al., 2009).

As a first approach, in vitro assays can predict the interaction of the nanomaterials with the body. However, the human body possesses compensation mechanisms when exposed to toxics and a huge disadvantage of this model is not to considered them. Moreover, they are less time consuming, more cost-effective, simpler and provide an easier control of the experimental conditions (Kroll et al., 2009; Fadeel et al., 2013b).

Their main drawback is the difficulty to reproduce all the complex interactions in the human body between sub-cellular levels, cells, organs, tissues and membranes. They use specific cells to achieve specific endpoints. In addition, in vitro assays cannot predict the physiopathological response of the human body when exposed to nanomaterials (Kroll et al., 2009; Fadeel et al., 2013b).

Another issue regarding the use of this approach is the possibility of interaction between nanomaterials and the reagents of the assay. It is likely that the reagents used in the in vitro assays interfere with the nanomaterial properties. High adsorption capacity, optical and magnetic properties, catalytic activity, dissolution, and acidity or alkalinity of the nanomaterials are some of the examples of properties that may promote this interaction (Kroll et al., 2009).

Many questions have been raised by the regulators related to the lack of consistency of the data produced by cytotoxicity assays. New assays for a correct evaluation of the nanomaterial toxicity are, thus, needed. In this context, new approaches have arisen, such as the in silico nanotoxicology approach. In silico methods are the combination of toxicology with computational tools and bio-statistical methods for the evaluation and prediction of toxicity. By using computational tools is possible to analyse more nanomaterials, combine different endpoints and pathways of nanotoxicity, being less time-consuming and avoiding all the ethical questions (Warheit, 2008; Raunio, 2011).

Quantitative structure-activity relationship models (QSAR) were one the first applications of computational tools applied in toxicology. QSAR models are based on the hypothesis that the toxicity of nanomaterials and their cellular fate in the body can be predicted by their characteristics, and different biological reactions are the result of physicochemical characteristics, such as size, shape, zeta potential, or surface charge, etc., gathered as a set of descriptors. QSAR aims at identifying the physicochemical characteristics which lead to toxicity, so as to provide alterations to reduce toxicology. A mathematical model is created, which allows liking descriptors and the biological activity (Rusyn and Daston, 2010; Winkler et al., 2013; Oksel et al., 2015).

Currently, toxigenomics is a new area of nanotoxicology, which includes a combination between genomics and nanotoxicology to find alterations in the gene, protein and in the expressions of metabolites (Rusyn et al., 2012; Fadeel et al., 2013a).

Hitherto, different risk assessment approaches have been reported. One of them is the DF4nanoGrouping framework, which concerns a functionality driven scheme for grouping nanomaterials based on their intrinsic properties, system dependent properties and toxicological effects (Arts et al., 2014, 2016). Accordingly, nanomaterials are categorized in four groups, including possible subgroups. The four main groups encompass (1) soluble, (2) biopersistent high aspect ratio, (3) passive, that is, nanomaterials without obvious biological effects and (4) active nanomaterials, that is, those demonstrating surface-related specific toxic properties. The DF4nanoGrouping foresees a stepwise evaluation of nanomaterial properties and effects with increasing biological complexity. In case studies that includes carbonaceous nanomaterials, metal oxide, and metal sulfate nanomaterials, amorphous silica and organic pigments (all nanomaterials having primary particle sizes smaller than 100 nm), the usefulness of the DF4nanoGrouping for nanomaterial hazard assessment has already been established. It facilitates grouping and targeted testing of nanomaterials, also ensuring that enough data for the risk assessment of a nanomaterial are available, and fostering the use of non-animal methods (Landsiedel et al., 2017). More recently, DF4nanoGrouping developed three structure-activity relationship classification, decision tree, models by identifying structural features of nanomaterials mainly responsible for the surface activity (size, specific surface area, and the quantum-mechanical calculated property lowest unoccupied molecular orbital), based on a reduced number of descriptors: one for intrinsic oxidative potential, two for protein carbonylation, and three for no observed adverse effect concentration (Gajewicz et al., 2018)

Keck and Mller also proposed a nanotoxicological classification system (NCS) (Figure 3) that ranks the nanomaterials into four classes according to the respective size and biodegradability (Mller et al., 2011; Keck and Mller, 2013).

Due to the size effects, this parameter is assumed as truly necessary, because when nanomaterials are getting smaller and smaller there is an increase in solubility, which is more evident in poorly soluble nanomaterials than in soluble ones. The adherence to the surface of membranes increases with the decrease of the size. Another important aspect related to size that must be considered is the phagocytosis by macrophages. Above 100 nm, nanomaterials can only be internalized by macrophages, a specific cell population, while nanomaterials below 100 nm can be internalized by any cell due to endocytosis. Thus, nanomaterials below 100 nm are associated to higher toxicity risks in comparison with nanomaterials above 100 nm (Mller et al., 2011; Keck and Mller, 2013).

In turn, biodegradability was considered a required parameter in almost all pharmaceutical formulations. The term biodegradability applies to the biodegradable nature of the nanomaterial in the human body. Biodegradable nanomaterials will be eliminated from the human body. Even if they cause some inflammation or irritation the immune system will return to the regular function after elimination. Conversely, non-biodegradable nanomaterials will stay forever in the body and change the normal function of the immune system (Mller et al., 2011; Keck and Mller, 2013).

There are two more factors that must be taken into account in addition to the NCS, namely the route of administration and the biocompatibility surface. When a particle is classified by the NCS, toxicity depends on the route of administration. For example, the same nanomaterials applied dermally or intravenously can pose different risks to the immune system.

In turn, a non-biocompatibility surface (NB) can activate the immune system by adsorption to proteins like opsonins, even if the particle belongs to the class I of the NCS (Figure 3). The biocompatibility (B) is dictated by the physicochemical surface properties, irrespective of the size and/or biodegradability. This can lead to further subdivision in eight classes from I-B, I-NB, to IV-B and IV-NB (Mller et al., 2011; Keck and Mller, 2013).

NCS is a simple guide to the evaluation of the risk of nanoparticles, but there are many other parameters playing a relevant role in nanotoxicity determination (Mller et al., 2011; Keck and Mller, 2013). Other suggestions encompass more general approaches, combining elements of toxicology, risk assessment modeling, and tools developed in the field of multicriteria decision analysis (Rycroft et al., 2018).

A forthcoming challenge in the pharmaceutical development is the scale-up and reproducibility of the nanomedicines. A considerable number of nanomedicines fail these requirements and, consequently, they are not introduced on the pharmaceutical market (Agrahari and Hiremath, 2017).

The traditional manufacturing processes do not create three dimensional medicines in the nanometer scale. Nanomedicine manufacturing processes, as already mentioned above, compromise top-down and bottom-down approaches, which include multiple steps, like homogenization, sonication, milling, emulsification, and sometimes, the use of organic solvents and further evaporation. In a small-scale, it is easy to control and achieve the optimization of the formulation. However, at a large scale it becomes very challenging, because slight variations during the manufacturing process can originate critical changes in the physicochemical characteristics and compromise the quality and safety of the nanomedicines, or even the therapeutic outcomes. A detailed definition of the acceptable limits for the CQA is very important, and these parameters must be identified and analyzed at the small-scale, in order to understand how the manufacturing process can change them: this will help the implementation of the larger scale. Thus, a deep process of understanding the critical steps and the analytical tools established for the small-scale will be a greatly help for the introduction of the large scale (Desai, 2012; Kaur et al., 2014; Agrahari and Hiremath, 2017).

Another requirement for the introduction of medicines in the pharmaceutical market is the reproducibility of every batch produced. The reproducibility is achieved in terms of physicochemical characterization and therapeutic purpose. There are specific ranges for the variations between different batches. Slight changes in the manufacturing process can compromise the CQA and, therefore, they may not be within a specific range and create an inter-batch variation (Desai, 2012; Kaur et al., 2014; Agrahari and Hiremath, 2017).

Over the last decades, nanomedicines have been successfully introduced in the clinical practice and the continuous development in pharmaceutical research is creating more sophisticated ones which are entering in clinic trials. In the European Union, the nanomedicine market is composed by nanoparticles, liposomes, nanocrystals, nanoemulsions, polymeric-protein conjugates, and nanocomplexes (Hafner et al., 2014). Table 2 shows some examples of commercially available nanomedicines in the EU (Hafner et al., 2014; Choi and Han, 2018).

In the process of approval, nanomedicines were introduced under the traditional framework of the benefit/risk analysis. Another related challenge is the development of a framework for the evaluation of the follow-on nanomedicines at the time of reference medicine patent expiration (Ehmann et al., 2013; Tinkle et al., 2014).

Nanomedicine comprises both biological and non-biological medical products. The biological nanomedicines are obtained from biological sources, while non-biological are mentioned as non-biological complex drugs (NBCD), where the active principle consists of different synthetic structures (Tinkle et al., 2014; Hussaarts et al., 2017; Mhlebach, 2018).

In order to introduce a generic medicine in the pharmaceutical market, several parameters need to be demonstrated, as described elsewhere. For both biological and non-biological nanomedicines, a more complete analysis is needed, that goes beyond the plasma concentration measurement. A stepwise comparison of bioequivalence, safety, quality, and efficacy, in relation to the reference medicine, which leads to therapeutic equivalence and consequently interchangeability, is required (Astier et al., 2017).

For regulatory purposes, the biological nanomedicines are under the framework set by European Medicines Agency (EMA) This framework is a regulatory approach for the follow-on biological nanomedicines, which include recommendations for comparative quality, non-clinical and clinical studies (Mhlebach et al., 2015).

The regulatory approach for the follow-on NBCDs is still ongoing. The industry frequently asks for scientific advice and a case-by-case is analyzed by the EMA. Sometimes, the biological framework is the base for the regulation of the NBCDs, because they have some features in common: the structure cannot be fully characterized and the in vivo activity is dependent on the manufacturing process and, consequently, the comparability needs to establish throughout the life cycle, as happens to the biological nanomedicines. Moreover, for some NBCDs groups like liposomes, glatiramoids, and iron carbohydrate complexes, there are draft regulatory approaches, which help the regulatory bodies to create a final framework for the different NBCDs families (Schellekens et al., 2014).

EMA already released some reflection papers regarding nanomedicines with surface coating, intravenous liposomal, block copolymer micelle, and iron-based nano-colloidal nanomedicines (European Medicines Agency, 2011, 2013a,b,c). These papers are applied to both new nanomedicines and nanosimilars, in order to provide guidance to developers in the preparation of marketing authorization applications.The principles outlined in these documents address general issues regarding the complexity of the nanosystems and provide basic information for the pharmaceutical development, non-clinical and early clinical studies of block-copolymer micelle, liposome-like, and nanoparticle iron (NPI) medicinal products drug products created to affect pharmacokinetic, stability and distribution of incorporated or conjugated active substances in vivo. Important factors related to the exact nature of the particle characteristics, that can influence the kinetic parameters and consequently the toxicity, such as the physicochemical nature of the coating, the respective uniformity and stability (both in terms of attachment and susceptibility to degradation), the bio-distribution of the product and its intracellular fate are specifically detailed.

After a nanomedicine obtains the marketing authorization, there is a long way up to the introduction of the nanomedicine in the clinical practice in all EU countries. This occurs because the pricing and reimbursement decisions for medicines are taken at an individual level in each member state of the EU (Sainz et al., 2015).

In order to provide patient access to medicines, the multidisciplinary process of Health Technology Assessment (HTA), is being developed. Through HTA, information about medicine safety, effectiveness and cost-effectiveness is generated so as support health and political decision-makers (Sainz et al., 2015).

Currently, pharmacoeconomics studies assume a crucial role previous to the commercialization of nanomedicines. They assess both the social and economic importance through the added therapeutic value, using indicators such as quality-adjusted life expectancy years and hospitalization (Sainz et al., 2015).

The EUnetHTA was created to harmonize and enhance the entry of new medicines in the clinical practice, so as to provide patients with novel medicines. The main goal of EUnetHTA is to develop decisive, appropriate and transparent information to help the HTAs in EU countries.

Currently, EUnetHTA is developing the Joint Action 3 until 2020 and the main aim is to define and implement a sustainable model for the scientific and technical cooperation on Health Technology Assessment (HTA) in Europe.

The reformulation of pre-existing medicines or the development of new ones has been largely boosted by the increasing research in nanomedicine. Changes in toxicity, solubility and bioavailability profile are some of the modifications that nanotechnology introduces in medicines.

In the last decades, we have assisted to the translation of several applications of nanomedicine in the clinical practice, ranging from medical devices to nanopharmaceuticals. However, there is still a long way toward the complete regulation of nanomedicines, from the creation of harmonized definitions in all Europe to the development of protocols for the characterization, evaluation and process control of nanomedicines. A universally accepted definition for nanomedicines still does not exist, and may even not be feasible at all or useful. The medicinal products span a large range in terms of type and structure, and have been used in a multitude of indications for acute and chronic diseases. Also, ongoing research is rapidly leading to the emergence of more sophisticated nanostructured designs that requires careful understanding of pharmacokinetic and pharmacodynamic properties of nanomedicines, determined by the respective chemical composition and physicochemical properties, which thus poses additional challenges in regulatory terms.

EMA has recognized the importance of the establishment of recommendations for nanomedicines to guide their development and approval. In turn, the nanotechnology methods for the development of nanomedicines bring new challenges for the current regulatory framework used.

EMA have already created an expert group on nanomedicines, gathering members from academia and European regulatory network. The main goal of this group is to provide scientific information about nanomedicines in order to develop or review guidelines. The expert group also helps EMA in discussions with international partners about nanomedicines. For the developer an early advice provided from the regulators for the required data is highly recommended.

The equivalence of complex drug products is another topic that brings scientific and regulatory challenges. Evidence for sufficient similarity must be gathered using a careful stepwise, hopefully consensual, procedure. In the coming years, through all the innovation in science and technology, it is expected an increasingly higher number of medicines based on nanotechnology. For a common understanding among different stakeholders the development of guidelines for the development and evaluation of nanomedicines is mandatory, in order to approve new and innovative nanomedicines in the pharmaceutical market. This process must be also carried out along with interagency harmonization efforts, to support rational decisions pertaining to scientific and regulatory aspects, financing and market access.

CV conceived the original idea and directed the work. SS took the lead in writing the manuscript. AP and JS helped supervise the manuscript. All authors provided critical feedback and helped shape the research, analysis and revision of the manuscript.

This work was financially supported by Fundao para a Cincia e a Tecnologia (FCT) through the Research Project POCI-01-0145-FEDER-016648, the project PEst-UID/NEU/04539/2013, and COMPETE (Ref. POCI-01-0145-FEDER-007440). The Coimbra Chemistry Center is supported by FCT, through the Project PEst-OE/QUI/UI0313/2014 and POCI-01-0145-FEDER-007630. This paper was also supported by the project UID/QUI/50006/2013LAQV/REQUIMTE.

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Adabi, M., Naghibzadeh, M., Adabi, M., Zarrinfard, M. A., Esnaashari, S., Seifalian, A. M., et al. (2017). Biocompatibility and nanostructured materials: applications in nanomedicine. Artif. Cells Nanomed. Biotechnol. 45, 833842. doi: 10.1080/21691401.2016.1178134

PubMed Abstract | CrossRef Full Text | Google Scholar

Agrahari, V., and Hiremath, P. (2017). Challenges associated and approaches for successful translation of nanomedicines into commercial products. Nanomedicine 12, 819823. doi: 10.2217/nnm-2017-0039

PubMed Abstract | CrossRef Full Text | Google Scholar

Albanese, A., Tang, P. S., and Chan, W. C. (2012). The effect of nanoparticle size, shape, and surface chemistry on biological systems. Annu. Rev. Biomed. Eng.14, 116. doi: 10.1146/annurev-bioeng-071811-150124

Read the original post:
Frontiers | Nanomedicine: Principles, Properties, and ...

Posted in Nano medicine | Comments Off on Frontiers | Nanomedicine: Principles, Properties, and …

[Full text] Relevance of PD-L1 Non-Coding Polymorphisms on the Prognosis of a Gene | PGPM – Dove Medical Press

Posted: February 19, 2021 at 1:44 am

Introduction

Lung cancer is one of the leading causes of cancer-related death globally.1 Although there are different types of lung cancer, non-small cell lung cancer (NSCLC) represents 85% of all primary lung tumors. NSCLC is a grim disease that is aggravated by the fact that patients normally either receive their diagnosis at advanced stages or present with recurrent disease after initial locoregional treatment.2 Over the last few decades, conventional chemotherapy, mainly platinum-based chemotherapy, used to be the only therapeutic option for those not eligible for radical intent treatment: a treatment with limited efficacy and very few long-term survivors (5-year overall survival less than 15%). Furthermore, these patients often lacked therapeutic options beyond first-line treatment.3

More recently, though, immune checkpoint molecules involved in tumor immune evasion were identified and immune checkpoint inhibitors (ICIs) were introduced in antitumor immunotherapy. This new therapeutic approach targets an inhibitory receptor, the programmed cell death-1 (PD-1) receptor, to assist the immune system in identifying and neutralizing malignant cells. However, tumor cells may evade the host immunosurveillance by expressing the programmed death-1-ligand 1 (PD-L1) as an adaptive, resistant mechanism to suppress this inhibitory receptor.4 Thus, because PD-L1 up-regulation by tumor cells can protect them from antitumor immune response, the blockade of PD-L1/PD-1 interactions has been recently selected for antitumor immune therapy.5 Agents targeting the PD-1/PD-L1 signaling pathway have shown promising responses in different types of cancer, including NSCLC. These results point to PD-L1 protein expression as a potential predictive marker for a successful blockade of PD-L1/PD-1 interactions.6 However, several challenges remain in producing robust evidence to support the use of this biomarker.

In this context, some studies with NSCLC patients have demonstrated that those with more than 50% PD-L1 positive tumor cells are non-responders to anti-PD-1/PD-L1 treatment. In contrast, others have shown that patients whose tumors do not express PD-L1 are good responders.79 To explain the controversies that affect PD-L1 expression, some studies have considered that the heterogeneity between axis expression and response to PD-1/PD-L1 treatment in NSCLC depends on other factors, such as more precise methods to investigate immune evasion mechanisms and the immune microenvironment, as well as greater knowledge on the immune checkpoint genomic profile and the genetic variants of the PD-L1 gene.1013

Some genetic variants have been shown to affect normal gene activation and transcriptional initiation, and hence influence the amount of mRNA and encoded protein in the cell.14 Non-coding variants also presumably affect genetic regulatory elements, since a majority of driver variants in cancer genomes occur in non-coding regions.15,16

Thus, several studies have investigated the association between PD-1 and PD-L1 genetic variants and the risk of various cancers, but their findings have yet failed to completely elucidate this question.17 A previous study suggested that PD-L1 polymorphism may predict chemotherapy response and survival rates in advanced-stage NSCLC patients after first-line paclitaxel-cisplatin.18 More recently, PD-L1 copy number variations, point mutations, and 3-UTR disruptions have been highlighted as genetic mechanisms of PD-L1 deregulation.19 Furthermore, previous research on Brazilian patients suggested that their ethnic background could account for their distinct cancers molecular profile, perhaps due to their characteristic genetic admixture, inherited from European, African, and Native American ancestors.2022

We hypothesize that PD-L1 non-coding genetic variants modulate the function of this immune checkpoint in NSCLC. To explore this issue, we investigated fifteen PD-L1 non-coding genetic variants using next-generation sequencing (NGS) in a Brazilian cohort, aiming to uncover the effect of their ethnic admixture on NSCLC. We also combined our analyses with an in-silico approach to predict the impact of these genetic variants on the disease. We evaluated the associations between PD-L1 protein expression level and clinicopathological characteristics, including the prognosis of NSCLC patients undergoing surgical resection, glimpsing the impact of genetic variants on post progression survival (PPS) and overall survival (OS). Herein, in this context, we intend to expand the existing literature on PD-L1 gene alterations at the genetic level and their impact on NSCLC in patients from different ethnicities, thus increasing the knowledge about the molecular basis of immunotherapy biomarkers.

In this retrospective multi-center study, we obtained archival formalin-fixed paraffin-embedded histologic tumor sections from 70 patients diagnosed with NSCLC (33 adenocarcinomas [ADC], 24 squamous cell carcinoma [SqCC] and 13 large cell carcinoma [LCC]) who underwent surgical resection between January 1, 1995, and December 31, 2015. Patients had been treated at the Hospital das Clnicas of the University of So Paulo Medical School (HC-FMUSP), at the Heart Institute of the University of So Paulo (INCOR), at the Cancer Institute of So Paulo (ICESP), and at the A.C. Camargo Cancer Center in So Paulo, Brazil.

All samples were histologically reviewed by lung pathologists who selected samples with at least 30% of lung cancer cells before nucleic acid extraction. The samples were classified using the 2017 International Association for the Study of Lung Cancer (IASLC) classification system.23 The clinicopathological features of patients were obtained from the medical records. The study was approved in accordance with the ethical standards of the responsible committee on human experimentation local (Research Ethics Committee of University of So Paulo Medical School - CAAE: 79769017.1.0000.5440; opinion number: 2.673.320) and with the 1964 Helsinki declaration. A waiver of the requirement for informed consent was obtained from committee, and to identity of the subjects under this retrospective analysis was omitted and anonymized.

We performed a Multiplex immunofluorescence (mIF) staining using methods that had been previously described and validated.24,25 Four-micrometer-thick consecutive TMA sections were stained using an automated staining system (BOND-RX; Leica Biosystems, Buffalo Grove, IL) to characterize PD-L1 (clone E1L3N, dilution 1:100; Cell Signaling Technology, Danvers, MA). The PD-L1 marker was stained with its respective fluorophore from the Opal 7 kit (catalogue #NEL797001KT; Akoya Biosciences/PerkinElmer, Waltham, MA). A complete validation using immunofluorescence (IF) allowed us to obtain a uniform, specific, and appropriate signal across all the channel; ie, a well-balanced staining pattern for the multiplex staining.24,25 We also defined and optimized the correct fluorophore signals between 10 and 30 counts of intensity to maintain good balance and similar thresholds of intensity across all antibodies. In parallel, to detect possible variations in staining and optimize the separation of the signal, positive and negative (autofluorescence) controls were included during the staining process to ensure that all the antibodies performed well together. Autofluorescence controls with an expected spectral resolution of 488nm were able to accurately remove the autofluorescence from all the label signals during the analysis. The stained slides were then scanned using a multispectral microscope, the Vectra Polaris 3.0 imaging system (Akoya Biosciences/PerkinElmer, Waltham, MA), under fluorescence conditions.

Multispectral images of tumor sections from each core were analyzed with inForm 2.2.1 (Akoya Biosciences/PerkinElmer, Waltham, MA) software Individual cells, which were defined by nuclei staining and identified by the InForm cell segmentation tool, were subjected to a phenotyping pattern-recognition learning algorithm to characterize co-localization of the various cell populations using panel labeling.26 The panel labeling was as follows: Malignant cells (MCs), with the AE1/AE3+ marker, including those with and without PD-L1 expression (AE1/AE3+ PD-L1+ and AE1/AE3+ PD-L1-, respectively). The individual cell phenotype report produced by the InForm software was processed using Excel 2010 (Microsoft. Houston, TX), and a final summary of the data, which contained the median of each individual phenotype (given as number of cells/mm2) and the percentage of macrophages and MCs expressing PD-L1, was created for statistical analysis. If the percentage of MCs or macrophages expressing PD-L1was greater than the median value, the PD-L1 expression was considered positive. If the percentage of macrophages or MCs expressing PD-L1 was lower than or equal to the median, the PD-L1 expression was considered negative.

Genomic DNA (gDNA) was extracted from frozen NSCLC tissue using the QIAamp DNA Mini Kit (Qiagen, Hilden, Germany) according to the manufacturers recommendations. DNA concentration was measured using the Qubit 3.0 Fluorometer (Invitrogen, Life Technologies, CA, USA). DNA integrity was assessed using the Bioanalyzer 2100 system (Agilent Technologies, CA, USA).

We performed a PD-L1 (CD274) full gene screening by deep targeted sequencing using the TruSeq Custom Amplicon Panel v1.5 kit (TSCAP, Illumina, SanDiego, CA) and the MiSeq platform (Illumina, SanDiego, CA). The DNA libraries were performed according to the manufacturers instructions and consisted of 150 bp paired-end reads (300 cycles).

We performed an NGS data analysis on the Molecular Genetics and Bioinformatics Laboratory of the Experimental Research Unit (UNIPEX) at the Medical School of So Paulo State University (FMB-UNESP). Sequencing quality was assessed by FastQC. Reads were aligned to the human genome (hg19, GRCh37) with BWA software, and SAM tools converted the alignment results to BAM format.27 Next, the mapped reads underwent variant calling for SNP with GATK command line tools, including HaplotypeCaller, SelectVariants, and VariantFiltration programs with default parameters. After the calling step, the variants were annotated using the VEP28 software. Coverage depth was a priori set at 100. Variants had to have >10 reads of position depth (PD) and/or >6 reads of allele depth (AD) and/or an AD/PD ratio of >0.05 and/or a population frequency higher than 1% (popfreq_all >0.01) were included in the study. Finally, variants were compared using ABraOM, a web-based public database of Brazilian genomic variants.29

Several tools were used to predict potential functional effects of SNPs on non-coding binding sites, such as splice sites and binding sites for transcription factors, exonic splicing enhancers (ESEs), and microRNA (miRNA). The impact of each genetic variant was assessed using VarSome30 an integrated search engine that allows access to several databases, forecasting tools, and publications on a single website. Variant pathogenicity was reported using an automatic variant classifier that evaluates each submitted variant according to guideline of the American College of Medical Genetics and Genomics (ACMG) and classifies it as either pathogenic, likely pathogenic, likely benign, benign or uncertain significance. Varsome predicts the pathogenicity of each variant through a DANN31 score, a methodology for scoring deleterious annotations of genetic variants using neural networks that results in a number ranging from 0 to 1. Higher DANN scores represent greater variant deleteriousness.31

Next, we applied the Genomic Evolutionary Rate Profiling (GERP)32,33 conservation score. This score is used to calculate the reduction of substitutions in a multi-species sequence alignment when compared to a neutral expectation. GERP scores >5.5 are strongly associated with a purifying selection. Mutations that occur at highly conserved sites in many species are assumed as harmful and therefore contribute to the genetic load within a species.

Finally, we used two tools, SNPinfo (FuncPred)34 and RegulomeDB,35 to track SNPs according to their functions. SNPinfo is a web server that helps researchers investigate SNPs in studies of genetic association and provide different pipelines for SNP selection, whereas RegulomeDB is an online composite database and prediction tool to annotate and prioritize potential regulatory variants from the human genome.35 RegulomeDB divides the variants into six categories: category 1 variants are likely to affect binding and are linked to the expression of a gene target, category 2 variants are likely to affect binding, category 3 variants are less likely to affect binding, and category 4, 5, and 6 variants have minimal binding evidence.35

The allelic and genotypic frequencies of the PD-L1 polymorphisms found in NSCLC were calculated by Hardy Weinberg equilibrium ([1 _ (hC 2H)]/2N, where h stands for a heterozygous genotype, H for homozygous genotype and N for the number of samples). Associations between polymorphisms, PD-L1 protein expression, and the clinicopathological parameters of NSCLC patients were investigated by Chi-square test. The prognostic value of each polymorphism was assessed by a survival analysis using the KaplanMeier method with the Log rank test for statistical significance. In addition, Coxs proportional hazards regression models were used in a multivariate analysis to test the association between SNPs and PPS and OS. PPS was considered as the period from tumor progression until death or last follow-up. OS was defined as the time from curative surgery to death or last date known to be alive. The statistical software program IBM SPSS (version 22; Armonk, NY, USA) performed all analyses. Differences were considered statistically significant at P<0.05.

Of the 70 patients included in the study, 33 presented with ADC (47.1%), 24 with SqCC (34.3%), and 13 with LCC (18.6%). The clinical characteristics by histologic types are summarized in Table 1. While SqCC cases were more frequent in males (81.8%), ADC cases were equally distributed between genders, and LCC cases were close to equal distribution (male 55.6%, female 44.4%). All histological types were more frequent in patients aged 63 years or younger. 8 patients reported a history of tobacco smoking in the ADC group (72.7%) and in the SqCC group (88.9%), versus 3 patients in the LCC group (50.0%). All the histological subtypes included advanced stages of disease (9 cases in ADC, 6 cases in SqCC, and 4 cases in LCC). Most of the patients had not received either chemotherapy (12 cases to ADC, 8 cases to SqCC, and 6 cases to LCC) or radiation therapy (18 cases to ADC, 10 cases to SqCC, and LCC) as adjuvant treatment. Malignant cells expressed PD-L1 above the median in 7 LCC cases (70.0%), the most relevant expression compared to the other two histological subtypes. The median follow-up was 66 (12144) months. None of the analysis revealed significant differences between histological types (P>0.05).

Table 1 Demographic and Clinicopathological Characteristics of 70 NSCLC Patients

All NSCLC patients who underwent surgical resection were successfully genotyped for fifteen PD-L1 SNPs: rs76805387T>C, rs4742098A>G, rs47946526A>G, rs10217310G>T, rs7864231G>A, rs41280725C>T, rs573692330A>G, rs1011769981G>A, rs41280723T>C, rs138135676T>C, rs4143815G>C, rs2297136G>A, rs148242519G>A, rs41303227C>T, and rs7041009G>A. Supplementary Table 1 shows the SNP identification numbers, allele and genotype frequencies, and P-value for HWE. Of the 15 SNPs studied, 11 were found to be monomorphic, whereas 4 SNPs, namely rs4742098, rs4143815, rs2297136, and rs7041009, were polymorphic in NSCLC. Monomorphic SNPs were excluded from further analysis. All the polymorphic SNPs were found to be in equilibrium (P>0.05) for HWE. The allele frequency of our cohort was compared to different populations in the 1000 Genomes Project (Supplementary Table 2).

We performed stratified analyses on the associations between clinical characteristics and the four PD-L1 polymorphisms with different genotypic distributions. Table 2 shows each SNP genotype frequency and their associated clinicopathological characteristics. Three of the four PD-L1 gene polymorphisms (rs4742098, rs4143815, and rs7041009) were significantly associated with relapse (P=0.01; P=0.05; P=0.02, respectively). For the rs4742098 variant, carriers of the G allele (AG or GG genotypes) were less likely to relapse (P=0.01). Similarly, for rs4143815, carriers of the alternative C allele (CG or CC genotypes) were also less likely to relapse (P=0.05). In rs7041009, however, carriers of the alternative allele A (AG or GG genotypes) were more likely to relapse (P=0.02). Moreover, GG genotype (reference) of rs7041009 showed a significant correlation with age, being more prevalent among younger patients (16 patients, or 69.6%), and status, being more prevalent among patients who were alive (11 patients, or 84.6%), compared to carriers of the A allele (P=0.02 and P<0.01, respectively). No statistical significance was observed in the association between rs2297136 genotypes and clinicopathological variants.

Table 2 Clinicopathological Characteristics of 70 NSCLC Patients Stratified by the PD-L1 Polymorphisms rs4742098, rs4143815, rs2297136 and rs7041009

The correlation between PD-L1 protein expression and PD-L1 gene polymorphisms are shown in Table 2. There were no statistically significant associations between PD-L1 protein expression in malignant cells and PD-L1 gene polymorphisms. In our cohort, the four PD-L1 gene polymorphisms were in non-coding regions and, apparently, cause no interference in PD-L1 protein expression in NSCLC malignant cells. However, when we correlated PD-L1 protein expression with histological subtype, we observed that the expression in malignant cells was above the median in 70% of patients with LLC, in contrast with 45.8% and 47.4% of patients with ADC and SqCC, respectively.

Our first statistical test examined the individual effect of patients characteristics to estimate statistical differences in survival using the KaplanMeier method (Table 3). Patients younger than 63 years showed increased OS, 111.62 vs 66.54 months in older patients (P=0.05). Choice of treatment was also an independent factor in diagnosis, with patients who did not receive radiotherapy presenting a better survival rate when compared with those who were treated with radiotherapy, 94.43 vs 12.00 months, respectively (P<0.01). Patients who presented disease recurrence had lower survival rates and poorer prognostic when compared with those who did not relapse, 48.23 vs 123.10 months, respectively (P<0.01).

Table 3 A Survival Analysis Conducted by the KaplanMeier Method Showing the Difference in the Means of the Log Rank Test According to the Optimal Upper and Lower Binary Cut-off Limits of Different Variables

Moreover, differences in the genotypes of PD-L1 polymorphisms seemed to also impact the prognosis of NSCLC patients. PD-L1 rs7041009, for instance, led to a statistically significant difference in OS (Figure 1), with carriers of the A allele of rs7041009 having lower OS than carriers of the GG genotype (reference), 59.00 vs 116.93 months, respectively (P<0.01).

Figure 1 KaplanMeier survival curve for PD-L1 rs7041009 G>A. A allele carriers (AG+AA) presented worse prognosis and a lower survival rate when compared to GG genotyped patients (P<0.01).

Next, using a univariate Cox Regression analysis, we were able to associate the following variables with a lower risk of death: the absence of radiotherapy treatment, relapse, and GG genotype of PDL1 rs7041009 (Table 4). However, after feeding these variables into a multivariate analysis, only the absence of radiotherapy treatment and relapse were considered to be independent factors for OS (HR 9.82, P=0.02; HR 6.15, P=0.04, respectively).

Table 4 Variables Associated with Overall Survival (OS) in 70 Patients Diagnosed with NSCLC. Univariate and Multivariate Analyses Employing a Cox Proportional Hazards Model

Then, we introduced the PD-L1 polymorphisms into the Cox model, controlling for radiotherapy treatment and tumor relapse. Of the four SNPs, only rs7041009 was identified as a co-dependent factor associated with radiotherapy and relapse. We thus inferred that patients with NSCLC who carried the A allele (AG/AA) presented a higher risk of relapse in the presence of radiotherapy, resulting in a poorer prognosis and decreased survival rates than patients who carried the rs7041009 GG genotype. In relapsed patients, we observed that the PD-L1 polymorphisms rs7041009 and rs4742098 might have an impact on PPS (Figure 2). Patients with the rs7041009 GG genotype had a higher PPS than those with the alternative A allele of rs7041009 (AG/AA), 110.98 vs 56.18 months, respectively (P<0.01); whereas, patients who carried the reference rs4742098 AA genotype had lower PPS than those who carried the alternative G allele of rs4742098 (AG/GG), 56.00 vs 115.71 months, respectively (P=0.02).

Figure 2 KaplanMeier survival curves estimating post-progression survival (PPS) in NSCLC patients according to PD-L1 polymorphisms. (A) KaplanMeier survival curve for rs7041009 G>A. A allele carriers (AG+AA) presented worse prognosis and a lower PPS rate when compared GG genotyped patients (P<0.01); (B) KaplanMeier survival curve for rs4742098 A>G. G allele carriers (AG+GG) had a higher PPS rate and better prognosis when compared AA genotyped patients (P=0.02).

The in silico analysis predicted the PD-L1 variants rs4742098 (c.*2635A>G), rs4143815 (c.*395G>C), rs2297136 (c.*93G>A), and rs7041009 (c.682+122G>A) to be benign (Table 5). Not only was the DANN score low for all four variants (0.8226, 0.6475, 0.7056, and 0.5428, respectively), but their GERP score was also lower than 5.5 (1.74, 2.38, 4.4, and 1.81, respectively), indicating that these variants are found in non-conserved positions and are unlikely to be harmful.

Table 5 List of the Selected Non-Coding SNP and the Tools Used to Study Them

SNPinfo predicted miRNA-binding function to be affected by two of these variants, namely rs2297136 and rs4143815. Rs2297136 was predicted to affect the binding function of hsa-miR-324-5p and hsa-miR-632, whereas rs4143815 was found to correlate with hsa-miR-1252, hsa-miR-1253, hsa-miR-539, hsa-miR-548, and hsa-miR-570 (Table 6). RegulomeDB was then used to complement the SNP analysis. Three of the four SNPs, rs4742098, rs4143815, and rs2297136, had a RegulomeDB score of 5, whereas rs7041009 had a score of 6, meaning that all four variants show minimal binding evidence (Table 5).

Table 6 List of the 3 UTR SNPs Analyzed in FuncPred and Their miRNA Motif

Lung cancer has a high mortality rate and lacks suitable markers for early diagnosis and prognosis. Thus, it is essential to detect the best potential biomarker out of the several genetic and protein markers. Fortunately for patients, the translational impact of such findings is rapidly increasing, and the stimulation of immune response by ICIs has emerged as a dramatic paradigm shift in the treatment of advanced tumors, mainly NSCLC.19,36 PD1/PDL1 monoclonal antibodies have shown potential efficacy in advanced squamous-cell and non-squamous NSCLC.37,38 However, despite the remarkable success achieved by immunotherapy so far, its effectiveness still seems to vary among cancer patients.19 The expression of PD-L1 on tumor cells remains the only recognized predictive factor for immunotherapy response in NSCLC patients; however, patients without PD-L1 expression on tumor cells may also respond to immunotherapy.3943 Based on these findings, the present study inferred that PD-L1 non-coding genetic variants could help predict the prognosis of patients with NSCLC and impact disease recurrence and OS.

In our cohort of 70 NSCLC specimens, we evaluated PD-L1 protein expression in malignant cells by PD-L1 multiplex immunofluorescence (mIF) assays, using the Cell Signaling E1L3N clone and image analysis, and investigated PD-L1 polymorphisms by NGS sequencing. This method resulted in the detection of high PD-L1 expression in LCC malignant cells when compared to other histological subtypes, suggesting that LCC patients may benefit from ICIs. As described by Shimoji et al,44 PD-L1 expression using the Cell Signaling E1L3N clone was significantly correlated with a consistent vimentin expression, increased Ki-67 labeling index and poor prognosis in ADC but not in SqCC. Other studies have reported that PD-L1 was detected at significantly higher frequencies in SqCC than in ADC of the lung.44,45 Cha et al46 found that PD-L1 expression using the SP142 clone was significantly associated with an ADC solid subtype histology, p53 aberrant expression, and poor prognosis.

We also assessed PD-L1 polymorphisms. Of the fifteen genetic variants genotyped, eleven were monomorphic. The four potential variants (rs4742098, rs4143815, rs2297136, and rs7041009) present in the non-coding region were correlated with the clinicopathological characteristics of the NSCLC patients and were submitted to an in silico analysis investigating their functional role. The MAF of the rs4742098, rs4143815, and rs7041009 polymorphisms was consistent with the genotype frequency among European and Mixed Americans populations present in The 1000 Genomes Project, whereas the G allele in the rs2297136 polymorphism was the main allele in our cohort to show racial differences.

When assessing patient prognosis, three of the four PD-L1 variants rs4742098, rs4143815, and rs7041009 were significantly associated with disease recurrence. Carriers of the G allele (individuals with the AG or GG genotypes) of rs4742098 were less likely to relapse compared to carriers of the homozygous AA genotype. Similar findings were published by Du and colleagues,47 who reported that the AG genotype differed from the AA genotype in terms of risk of NSCLC recurrence. In the case of rs4143815G, patients with the alternative C allele were less likely to relapse in our study, in agreement with Nomizo et als report.48 In their study, the authors even suggested that this polymorphism might be a biomarker for nivolumab efficacy.48 Finally, in our study, for rs7041009G>A, carriers of the alternative G allele were more likely to exhibit relapse. The rs7041009 GG genotype also showed a significant correlation with age, being more present in younger patients, and with status, being more present in patients who are alive, compared to carriers of the A allele. Rs7041009 (c.682+122G>A) is located at position 2377 in intron 4 of the PD-L1 gene. However, little is known about the exact function of this genetic variation, except that it is located near the transcription factor binding site.

Our cohort showed a significant association between these PD-L1 polymorphisms and OS in NSCLC. 18 Patients presenting the GG genotype of rs7041009 benefited from longer OS. In addition, our findings indicated that both the rs7041009G and rs4742098G polymorphisms were significantly related to a longer PPS. The clinical impacts of PD-L1 variants had also been investigated by previous studies. Zhao et al49 suggested that patients with the GG genotype of another PD-L1 polymorphism (rs822336) had worse disease-free survival and OS in a Chinese patient population.

Further contribution was provided in that respect by Lee et al50 who demonstrated that rs4143815 and rs2297136 were significantly associated with clinical outcomes after chemotherapy. Of 379 patients with NSCLC treated with first-line paclitaxelcisplatin chemotherapy, those carrying the rs4143815 G allele responded better to chemotherapy and had gains in overall survival. In our study, however, the polymorphism rs2297136 showed no significant association with clinical outcome, a finding that was corroborated by Zhao et al.49 This difference might be explained by the heterogeneity of patients enrolled in each study, and further research is needed to settle this question.

In our study, we were unable to find any statistical significance between the rs4742098, rs4143815, rs2297136, and rs7041009 genotypes and PD-L1 protein expression in malignant cells. However, recent data have helped to shed light on the impact of PD-L1 genetics on PD-L1 expression, though the existing results remain controversial. Recently, Tao and colleagues51 showed that rs4143815 and rs10815225 in the PD-L1 gene contributed to PD-L1 overexpression in gastric cancer. So far, Lee et al18 have conducted the largest study on PD-L1 polymorphisms and PD-L1 expression in NSCLC. The authors showed that rs822336C, rs822337A and rs4143815G were associated with worse OS in NSCLC patients but found no significant correlation between PD-L1 expression and the genotypes of these polymorphisms. Krawczyk et al52 demonstrated that carriers of the rs822335 CC genotype were predisposed to higher expression of the PD-L1 protein in NSCLC tumor cells, whereas rs822336 had no effect on PD-L1 expression in these cells. Their results are consistent with our findings, but future research is needed to clarify remaining confounders.

In our study, using in silico approaches, we report that rs4742098, rs4143815, rs2297136, and rs7041009 can be considered benign variants. However, little is known about the effect that mutations in conserved non-coding regions might have on fitness and how many of them are present in the human genome as deleterious polymorphisms. Moreover, rs2297136 was predicted to affect the miRNA-binding function of hsa-miR-324-5p and hsa-miR-632, whereas the variant rs4143815 was found to correlate with hsa-miR-1252, hsa-miR-1253, hsa-miR-539, hsa-miR-548, and hsa-miR-570. In this context, others have reported that variants in the 3UTR region of the PD-L1 gene can affect the interaction of miRNAs, possibly resulting in PD-L1 underexpression.53 Therefore, additional studies are necessary to validate our findings. However, there are limitations to our analysis. We did not perform a casecontrol approach and our cohort comprises a relatively small sample size. Nonetheless, to our knowledge, our research on the effect of the rs7041009 polymorphism of PD-L1 gene on NSCLC patients is unique.

We believe this study is also the first to evaluate variants in the non-coding region of the PD-L1 in Brazilian patients with NSCLC, since most studies of PD-L1 polymorphisms have been conducted in Asian patients. Thus, we consider this exploratory study as a pioneer in the understanding of PD-L1 polymorphisms in a genetic admixed population.

We appreciate all subjects who participated in this study and the Illumina members for assistance with the initial runs.

This study was supported by the So Paulo Research Foundation, FAPESP (grant numbers 2013/14277-4 and 2018/20403-6).

The authors report no conflicts of interest related to this work.

1. Shankar A, Saini D, Dubey A, et al. Feasibility of lung cancer screening in developing countries: challenges, opportunities and way forward. Transl Lung Cancer Res. 2019;8(Suppl S1):S106S121. doi:10.21037/tlcr.2019.03.03

2. Berghmans T, Durieux V, Hendriks LEL, Dingemans A-M. Immunotherapy: from Advanced NSCLC to Early Stages, an Evolving Concept. Front Med. 2020;7:90. doi:10.3389/fmed.2020.00090

3. Bylicki O, Paleiron N, Rousseau-Bussac G, Chouad C. New PDL1 inhibitors for non-small cell lung cancer: focus on pembrolizumab. Onco Targets Ther. 2018;11:40514064. doi:10.2147/OTT.S154606

4. Yeo M-K, Choi S-Y, Seong I-O, Suh K-S, Kim JM, Kim K-H. Association of PD-L1 expression and PD-L1 gene polymorphism with poor prognosis in lung adenocarcinoma and squamous cell carcinoma. Hum Pathol. 2017;68:103111. doi:10.1016/j.humpath.2017.08.016

5. Francisco LM, Salinas VH, Brown KE, et al. PD-L1 regulates the development, maintenance, and function of induced regulatory T cells. J Exp Med. 2009;206(13):30153029. doi:10.1084/jem.20090847

6. Lantuejoul S, Damotte D, Hofman V, Adam J. Programmed death ligand 1 immunohistochemistry in non-small cell lung carcinoma. J Thorac Dis. 2019;11(Suppl S1):S89S101. doi:10.21037/jtd.2018.12.103

7. Aggarwal C, Abreu DR, Felip E, et al. Prevalence of PD-L1 expression in patients with non-small cell lung cancer screened for enrollment in KEYNOTE-001, 010, and 024. Ann Oncol. 2016;27(6):359378. doi:10.1093/annonc/mdw378.14

8. Yu H, Boyle TA, Zhou C, Rimm DL, Hirsch FR. PD-L1 Expression in Lung Cancer. J Thorac Oncol. 2016;11(7):964975. doi:10.1016/j.jtho.2016.04.014

9. Reck M, Rodrguez-Abreu D, Robinson AG, et al. Pembrolizumab versus Chemotherapy for PD-L1Positive NonSmall-Cell Lung Cancer. N Engl J Med. 2016;375(19):18231833. doi:10.1056/NEJMoa1606774

10. Marwitz S, Scheufele S, Perner S, Reck M, Ammerpohl O, Goldmann T. Epigenetic modifications of the immune-checkpoint genes CTLA4 and PDCD1 in non-small cell lung cancer results in increased expression. Clin Epigenetics. 2017;9(1):51. doi:10.1186/s13148-017-0354-2

11. Chen L, Gibbons DL, Goswami S, et al. Metastasis is regulated via microRNA-200/ZEB1 axis control of tumour cell PD-L1 expression and intratumoral immunosuppression. Nat Commun. 2014;5(1):5241. doi:10.1038/ncomms6241

12. Ma Y, Adjemian S, Mattarollo SR, et al. Anticancer chemotherapy-induced intratumoral recruitment and differentiation of antigen-presenting cells. Immunity. 2013;38(4):729741. doi:10.1016/j.immuni.2013.03.003

13. Mazzaschi G, Madeddu D, Falco A, et al. Low PD-1 Expression in Cytotoxic CD8 + Tumor-Infiltrating Lymphocytes Confers an Immune-Privileged Tissue Microenvironment in NSCLC with a Prognostic and Predictive Value. Clin Cancer Res. 2018;24(2):407419. doi:10.1158/1078-0432.CCR-17-2156

14. de Vooght KMK, van Wijk R, van Solinge WW. Management of Gene Promoter Mutations in Molecular Diagnostics. Clin Chem. 2009;55(4):698708. doi:10.1373/clinchem.2008.120931

15. Cuykendall TN, Rubin MA, Khurana E. Non-coding genetic variation in cancer. Curr Opin Syst Biol. 2017;1:915. doi:10.1016/j.coisb.2016.12.017

16. Amlie-Wolf A, Tang M, Way J, et al. Inferring the Molecular Mechanisms of Noncoding Alzheimers Disease-Associated Genetic Variants. J Alzheimers Dis. 2019;72(1):301318. doi:10.3233/JAD-190568

17. Hashemi M, Karami S, Sarabandi S, et al. Association between PD-1 and PD-L1 Polymorphisms and the Risk of Cancer: A Meta-Analysis of Case-Control Studies. Cancers. 2019;11(8):1150. doi:10.3390/cancers11081150

18. Lee SY, Jung DK, Choi JE, et al. Functional polymorphisms in PD-L1 gene are associated with the prognosis of patients with early stage non-small cell lung cancer. Gene. 2017;599:2835. doi:10.1016/j.gene.2016.11.007

19. Fabrizio FP, Trombetta D, Rossi A, Sparaneo AA, Castellana S, Muscarella LA. Gene code CD274/PD-L1: from molecular basis toward cancer immunotherapy. Ther Adv Med Oncol. 2018;10:1758835918815598. doi:10.1177/1758835918815598

20. Araujo LH, Baldotto CA, Castro JGD, et al. Lung cancer in Brazil. J Bras Pneumol. 2018;44(1):5564. doi:10.1590/s1806-37562017000000135

21. de Melo AC, de S VK, Sternberg C, et al. Mutational Profile and New IASLC/ATS/ERS Classification Provide Additional Prognostic Information about Lung Adenocarcinoma: A Study of 125 Patients from Brazil. Oncology. 2015;89(3):175186. doi:10.1159/000376552

22. de S VK, Coelho JC, Capelozzi VL, de Azevedo SJ. Lung cancer in Brazil: epidemiology and treatment challenges.. Lung Cancer. 2016;7:141148. doi:10.2147/LCTT.S93604

23. Goldstraw P, Chansky K, Crowley J, et al. The IASLC Lung Cancer Staging Project: proposals forRevision of the TNM Stage Groupings in the Forthcoming (Eighth) Edition of the TNM Classification for Lung Cancer. J Thorac Oncol. 2016;11(1):3951. doi:10.1016/j.jtho.2015.09.009

24. Parra ER, Uraoka N, Jiang M, et al. Validation of multiplex immunofluorescence panels using multispectral microscopy for immune-profiling of formalin-fixed and paraffin-embedded human tumor tissues. Sci Rep. 2017;7(1):13380. doi:10.1038/s41598-017-13942-8

25. Parra ER, Jiang M, Machado-Rugolo J, et al. Variants in Epithelial-Mesenchymal Transition and Immune Checkpoint Genes Are Associated With Immune Cell Profiles and Predict Survival in NonSmall Cell Lung Cancer. Arch Pathol Lab Med. 2020;144(10):12341244. doi:10.5858/arpa.2019-0419-OA

26. Gorris MAJ, Halilovic A, Rabold K, et al. Eight-Color Multiplex Immunohistochemistry for Simultaneous Detection of Multiple Immune Checkpoint Molecules within the Tumor Microenvironment. J Immunol. 2018;200(1):347354. doi:10.4049/jimmunol.1701262

27. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25(14):17541760. doi:10.1093/bioinformatics/btp324

28. McLaren W, Gil L, Hunt SE, et al. The Ensembl Variant Effect Predictor.. Genome Biol. 2016;17(1):122. doi:10.1186/s13059-016-0974-4

29. Naslavsky MS, Yamamoto GL, de Almeida TF, et al. Exomic variants of an elderly cohort of Brazilians in the ABraOM database. Hum Mutat. 2017;38(7):751763. doi:10.1002/humu.23220

30. Kopanos C, Tsiolkas V, Kouris A, et al. VarSome: the human genomic variant search engine. Bioinformatics. 2019;35(11):19781980. doi:10.1093/bioinformatics/bty897

31. Quang D, Chen Y, Xie X. DANN: a deep learning approach for annotating the pathogenicity of genetic variants. Bioinformatics. 2015;31(5):761763. doi:10.1093/bioinformatics/btu703

32. Davydov EV, Goode DL, Sirota M, Cooper GM, Sidow A, Batzoglou S. Identifying a high fraction of the human genome to be under selective constraint using GERP++. PLoS Comput Biol. 2010;6(12):e1001025. doi:10.1371/journal.pcbi.1001025

33. Cooper GM, et al. Distribution and intensity of constraint in mammalian genomic sequence. Genome Res. 2005;15(7):901913. doi:10.1101/gr.3577405

34. Xu Z, Taylor JA. SNPinfo: integrating GWAS and candidate gene information into functional SNP selection for genetic association studies. Nucleic Acids Res. 2009;37(suppl_2):W600W605. doi:10.1093/nar/gkp290

35. Boyle AP, Hong EL, Hariharan M, et al. Annotation of functional variation in personal genomes using RegulomeDB. Genome Res. 2012;22(9):17901797. doi:10.1101/gr.137323.112

36. Nishino M, Ramaiya NH, Hatabu H, Hodi FS. Monitoring immune-checkpoint blockade: response evaluation and biomarker development. Nat Rev Clin Oncol. 2017;14(11):655668. doi:10.1038/nrclinonc.2017.88

37. Borghaei H, Paz-Ares L, Horn L, et al. Nivolumab versus Docetaxel in Advanced Nonsquamous NonSmall-Cell Lung Cancer. N Engl J Med. 2015;373(17):16271639. doi:10.1056/NEJMoa1507643

38. Brahmer J, Reckamp KL, Baas P, et al. Nivolumab versus Docetaxel in Advanced Squamous-Cell NonSmall-Cell Lung Cancer. N Engl J Med. 2015;373(2):123135. doi:10.1056/NEJMoa1504627

39. Yi C, He Y, Xia H, Zhang H, Zhang P.

Review and perspective on adjuvant and neoadjuvant immunotherapies in NSCLC. Onco Targets Ther. 2019;12:73297336. doi:10.2147/OTT.S218321

40. Horn L, Spigel DR, Vokes EE, et al. Nivolumab Versus Docetaxel in Previously Treated Patients With Advanced NonSmall-Cell Lung Cancer: two-Year Outcomes From Two Randomized, Open-Label, Phase III Trials (CheckMate 017 and CheckMate 057). J Clin Oncol. 2017;35(35):39243933. doi:10.1200/JCO.2017.74.3062

41. Antonia SJ, Villegas A, Daniel D, et al. Durvalumab after Chemoradiotherapy in Stage III NonSmall-Cell Lung Cancer. N Engl J Med. 2017;377(20):19191929. doi:10.1056/NEJMoa1709937

42. Fehrenbacher L, von Pawel J, Park K, et al. Updated Efficacy Analysis Including Secondary Population Results for OAK: A Randomized Phase III Study of Atezolizumab versus Docetaxel in Patients with Previously Treated Advanced NonSmall Cell Lung Cancer. J Thorac Oncol. 2018;13(8):11561170. doi:10.1016/j.jtho.2018.04.039

43. Gandhi L, Rodrguez-Abreu D, Gadgeel S, et al. Pembrolizumab plus Chemotherapy in Metastatic NonSmall-Cell Lung Cancer. N Engl J Med. 2018;378(22):20782092. doi:10.1056/NEJMoa1801005

44. Shimoji M, Shimizu S, Sato K, et al. Clinical and pathologic features of lung cancer expressing programmed cell death ligand 1 (PD-L1). Lung Cancer. 2016;98:6975. doi:10.1016/j.lungcan.2016.04.021

45. Takada K, Toyokawa G, Okamoto T, et al. A Comprehensive Analysis of Programmed Cell Death Ligand-1 Expression With the Clone SP142 Antibody in NonSmall-Cell Lung CancerPatients. Clin Lung Cancer. 2017;18(5):572582. doi:10.1016/j.cllc.2017.02.004

46. Cha YJ, Kim HR, Lee CY, Cho BC, Shim HS. Clinicopathological and prognostic significance of programmed cell death ligand-1 expression in lung adenocarcinoma and its relationship with p53 status. Lung Cancer. 2016;97:7380. doi:10.1016/j.lungcan.2016.05.001

47. Du W, Zhu J, Chen Y, et al. Variant SNPs at the microRNA complementary site in the B7-H1 3-untranslated region increase the risk of non-small cell lung cancer. Mol Med Rep. 2017;16(3):26822690. doi:10.3892/mmr.2017.6902

48. Nomizo T, Ozasa H, Tsuji T, et al. Clinical Impact of Single Nucleotide Polymorphism in PD-L1 on Response to Nivolumab for Advanced Non-Small-Cell Lung Cancer Patients. Sci Rep. 2017;7(1):45124. doi:10.1038/srep45124

49. Zhao M, Zhang J, Chen S, Wang Y, Tian Q.

Influence of Programmed Death Ligand-1-Gene Polymorphism rs822336 on the Prognosis and Safety of Postoperative Patients with NSCLC Who Received Platinum-Based Adjuvant Chemotherapy. Cancer Manag Res. 2020;12:67556766. doi:10.2147/CMAR.S255072

50. Lee SY, Jung DK, Choi JE, et al. PD-L1 polymorphism can predict clinical outcomes of non-small cell lung cancer patients treated with first-line paclitaxel-cisplatin chemotherapy. Sci Rep. 2016;6(1):25952. doi:10.1038/srep25952

51. Tao L-H, Zhou X-R, Li F-C, et al. A polymorphism in the promoter region of PD-L1 serves as a binding-site for SP1 and is associated with PD-L1 overexpression and increased occurrence of gastric cancer. Cancer Immunol Immunother. 2017;66(3):309318. doi:10.1007/s00262-016-1936-0

52. Krawczyk P, Grenda A, Wojas-Krawczyk K, et al. PD-L1 gene copy number and promoter polymorphisms regulate PD-L1 expression in tumor cells of non-small cell lung cancer patients. Cancer Genetics. 2019;237:1018. doi:10.1016/j.cancergen.2019.06.001

See the original post here:
[Full text] Relevance of PD-L1 Non-Coding Polymorphisms on the Prognosis of a Gene | PGPM - Dove Medical Press

Posted in Molecular Genetics | Comments Off on [Full text] Relevance of PD-L1 Non-Coding Polymorphisms on the Prognosis of a Gene | PGPM – Dove Medical Press

Catamaran Bio Launches with $42 Million Financing to Develop OfftheShelf CAR-NK Cell Therapies to Treat Solid Tumors – BioSpace

Posted: November 25, 2020 at 11:54 pm

In assembling the founding team at Catamaran, we saw an opportunity to pioneer a highly differentiated approach to develop allogeneic cell therapies using CAR-NK cells, said Houman Ashrafian, Managing Partner, SV Health Investors and a founder of Catamaran. To date, the success of autologous CAR T-cell therapies in hematological malignancies has opened the door to the breakthrough potential of cell therapies for cancer, and Catamaran is now well positioned to improve upon this groundwork by developing off-the-shelf CAR-NK cell therapies capable of reaching solid tumors.

A novel approach to developing off-the-shelf cell therapies to address solid tumors

Catamarans TAILWIND Platform integrates proprietary capabilities to create novel, allogeneic CARNK cell therapies by harnessing the natural cancer-fighting properties of natural killer (NK) cells and enhancing them with the power of synthetic biology and innovative NK cell engineering and manufacturing. With the TAILWIND Platform, CAR-NK cells are programmed with NK cell-specific CAR architectures and potency-boosting switches to neutralize the hostile tumor microenvironment and enable efficacy against diverse cancer types, especially solid tumors. Additionally, the TAILWIND Platform includes proprietary, non-viral NK cell engineering technology for efficient modification of NK cells with customized genetic programs enabled by synthetic biology. Catamarans CAR-NK cell therapies use healthy donor cells that are engineered and manufactured for offtheshelf use, unlike current CAR-T cell therapies that use a patients own genetically modified T cells and require a customized, multi-week manufacturing process.

Catamaran is focused on expanding the frontier of cell therapies to treat solid tumors and provide transformative benefit to cancer patients. We are doing this by creating allogeneic cell therapies that harness the innate cancer-fighting power of NK cells and enhancing them with new biologically-powerful attributes from our leading-edge technologies all originating from our custom-built TAILWIND Platform for designing, engineering and manufacturing off-the-shelf CAR-NK cell therapies, said Vipin Suri, PhD, MBA, Chief Scientific Officer of Catamaran.

During Catamarans stealth period, the start-up team assembled key components of the TAILWIND Platform and related intellectual property, including a set of potency-boosting cellular switches to enable therapeutic action in the immunosuppressive tumor microenvironment of solid tumors, and it generated early proof of concept using a non-viral transposon system to efficiently deliver large genetic cargos into NK cells. Based on this early work, the company has rapidly advanced two lead CAR-NK cell therapy programs to lead optimization stage.

With its holistic and cutting-edge approach, Catamaran stands out in the rapidly-evolving NK cell field with a platform that addresses the full complement of capabilities necessary to develop CAR-NK cell therapies, while focusing on the high-impact technologies of synthetic biology and innovative gene delivery systems that can enable these new cell therapies to offer extraordinary value in the field of cancer treatment, said Maina Bhaman, Partner, Sofinnova Partners.

Scientific founders and leadership team

Catamarans scientific founders are pioneers in NK cell biology, engineering, manufacturing and clinical application and are proven innovators in the cell therapy field:

Additional founders of Catamaran are Kevin Pojasek, PhD, and Tim Harris, PhD, through their roles as venture partners with SV Health Investors.

The leadership team at Catamaran Bio has deep expertise in cell therapy research and product development, and the team includes: Vipin Suri, PhD, MBA, Chief Scientific Officer, who has more than 20 years of biopharmaceutical experience, including as a co-founder of Obsidian Therapeutics and Serien (formerly Raze) Therapeutics, and earlier in R&D roles at GSK, Pfizer and Wyeth; Mark Boshar, JD, Chief Operating Officer, who has more than 25 years of leadership experience spanning legal, business development, financings and operations for biotechnology companies, including as VP, Legal Affairs at Rubius Therapeutics, Associate General Counsel at Millennium Pharmaceuticals, a senior advisor to a range of venture-backed start-up companies, and earlier as a life sciences attorney with WilmerHale; Chris Carpenter, MD, PhD, Chief Medical Officer, who has 20 years of clinical and laboratory experience in oncology, including as CMO of Rubius Therapeutics, SVP and Head of Cancer Epigenetics Discovery at GSK, and roles at Merck and Harvard Medical School/Beth Israel Deaconess Medical Center; Celeste Richardson, PhD, Senior VP of Research, who has 16 years of experience in research and drug discovery in biotechnology and pharmaceutical companies, including at Obsidian Therapeutics and Novartis; and Bharat Reddy, PhD, MPhil, MA, Senior Director of Business Development, who has served as director of business development at bluebird bio, as well as roles at SV Health Investors and ClearView Healthcare Partners.

Catamaran is positioned to open up new territory for cancer treatments with highly potent CAR-NK cell therapies, and we are confident in the experienced leadership team and the scientific expertise that is propelling the companys research and development, said Caroline Gaynor, Principal, Lightstone Ventures.

Concurrent with the Series A financing, Maina Bhaman of Sofinnova Partners, Caroline Gaynor of Lightstone Ventures and Rob Woodman of Takeda Ventures join Houman Ashrafian and Kevin Pojasek on the Catamaran board of directors.

About Catamaran Bio

Catamaran Bio is developing novel, off-the-shelf CAR-NK cell therapies designed to treat a broad range of cancers, including solid tumors. Our proprietary capabilities enable us to harness the natural cancer-fighting properties of NK cells and enhance and tailor their effectiveness with the power of synthetic biology and innovative non-viral cell engineering. We are using our TAILWINDTM Platform, an integrated suite of technologies, to specifically address the end-to-end methods of engineering, processing and manufacturing NK cells and rapidly advance our pipeline of CAR-NK cell therapy programs.

Our team combines experienced biopharmaceutical leadership with founding scientists who are pioneers in NK cell biology, engineering, manufacturing and clinical application. Catamaran is backed by leading financial and corporate investors, including SV Health Investors, Sofinnova Partners, Lightstone Ventures, Takeda Ventures and Astellas Venture Management. For more information, please visit http://www.catamaranbio.com and follow us on LinkedIn and @CatamaranBio on Twitter.

View source version on businesswire.com: https://www.businesswire.com/news/home/20201123005485/en/

Read the original here:
Catamaran Bio Launches with $42 Million Financing to Develop OfftheShelf CAR-NK Cell Therapies to Treat Solid Tumors - BioSpace

Posted in Cell Therapy | Comments Off on Catamaran Bio Launches with $42 Million Financing to Develop OfftheShelf CAR-NK Cell Therapies to Treat Solid Tumors – BioSpace

Ontario Announces First Phase of Research Projects to Fight COVID-19 – Government of Ontario News

Posted: May 24, 2020 at 12:47 am

Ontario Announces First Phase of Research Projects to Fight COVID-19Ontario-Based Solutions Contribute to the Global Effort against the Outbreak

Ontario is funding the following research on preventing, detecting and treating COVID-19. These projects focus on important areas of research, including vaccine development, diagnostics, drug trials and development, and social sciences.

A Randomized Open-Label Trial of CONvalescent Plasma for Hospitalized Adults with Acute COVID-19 Respiratory Illness (CONCOR-1)Donald Arnold, Principal InvestigatorMcMaster University

CONCOR-1 is a clinical trial that will collect blood plasma from individuals who have recovered from COVID-19, known as COVID-19 convalescent plasma. Convalescent plasma contains COVID-19 antibodies, proteins that help fight the virus. Convalescent plasma will be injected into patients currently fighting the infection, to test whether this is an effective treatment for the virus. This clinical trial will enrol patients 16 years of age and older admitted to hospital with COVID-19 and who require supplemental oxygen for respiratory illness.

Partners include 60 hospitals across Canada and three hospitals in New York City, the Canadian Blood Services and Hma-Qubec and the New York Blood Center.

Research and Deployment of Rapid High-Throughput Diagnostic Testing for COVID-19Marek Smieja, Principal InvestigatorSt Joseph's Healthcare Hamilton

This project will increase Ontario's COVID-19 testing capacity by deploying robotic liquid handling technology, specimen pooling, and efficient sample preparation, while reducing biological risk and ensuring reliable results. The Disease Diagnostics & Development group in the Research Institute of St Joe's Hamilton (RSJH) is collaborating with the Hamilton Regional Laboratory Medicine Program (HRLMP) and other clinical laboratories across the province to quickly develop, validate, and deliver high-throughput, COVID-19 testing, with the goal of testing up to 6,000 samples per lab daily.

Assay Development for SARS-CoV-2 Sero-SurveillanceJennifer Gommerman, Principal InvestigatorUniversity of Toronto

This study will provide a better understanding of the immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes COVID-19. This approach aims to measure the level and/or types of antibodies induced by SARS-CoV-2 infection in the blood of acute and convalescent patients. In addition, measuring these antibodies in the saliva of asymptomatic infected subjects identified through contact tracing will provide insights into what the early immune response to the virus looks like, and how this may correlate with clinical outcome. This knowledge, as well as the development of a robust serosurveillance platform, represents a powerful weapon in our fight against COVID-19.

Multivalent Antibody Scaffold to Deliver an Exceptionally Potent and Broad Antiviral Against SARS-CoV-2Jean-Philippe Julien, Principal InvestigatorThe Hospital for Sick Children

This project has the potential to develop a unique antibody-based molecule for protection and treatment against COVID-19. Molecular technology will allow these researchers to decipher the vulnerabilities of the virus with the goal of developing a potent and broad antiviral that neutralizes SARS-CoV-2 and prevents associated COVID-19 symptoms.

Developing Prophylactic Virus-Vectored Vaccines for COVID-19Byram Bridle, Leonardo Susta and Sarah Wootton (Co-Principal Investigators, University of Guelph); Darwyn Kobasa, National Microbiology Laboratory, Public Health Agency of Canada (Collaborator) University of Guelph

This research aims to develop a vaccination strategy for COVID-19. By developing avian avulavirus (AAvV-1) and adenovirus viral-vectored vaccines expressing the SARS-CoV-2 spike protein as a target antigen, researchers will test these vaccines in mice to identify a way to induce robust protective mucosal (respiratory, gastrointestinal and urogenital tract) and systemic immunity. Mucosal immunity plays a significant role in preventing pathogens from getting into the body. Systemic immunity clears any pathogens that bypass mucosal barriers. After optimization, these vaccines will be evaluated in a hamster challenge model at the National Microbiology Laboratory in Winnipeg.

The RAPID COVID Study - Application of Point-of-Care COVID-19 Testing to Optimize Patient Care, Resource Allocation and Safety for Frontline StaffDerek So, Principal InvestigatorUniversity of Ottawa Heart Institute

This study will determine the role of point-of-care testing (POC) as a tool to improve care of COVID-19 patients and conserve resources. A major obstacle facing hospitals during the COVID-19 outbreak is the inability to quickly diagnose who is infected with the virus. Delayed test results could mean that patients, who ultimately test negative, are treated for days utilising resources that could be better deployed elsewhere. An immediate diagnosis of COVID-19 among carriers could provide more expedient treatment, prevent clinical deterioration and help health care workers avoid unnecessary risk of exposure.

In collaboration with Spartan Biosciences, which has developed a novel point-of-care 45-minute bedside COVID-19 test, and a team of specialists from six centres in Ontario, this research will evaluate the efficacy of POC testing to determine when, how and to who it can be applied.

A Prospective, Observational Research Study on the Diagnosis of COVID-19 Infection from Stool Samples of Children and AdultsNikhil Pai, Jeff Pernica, Marek Smieja (Co-Principal Investigators)McMaster University

Through the development and use of a novel test to diagnose COVID-19 from stool samples, this team will assess up to 4,500 stool samples collected from outpatient clinics, emergency departments and inpatient wards across eight major Hamilton region hospitals and clinics. This work will improve COVID-19 disease detection in children and adults who lack respiratory symptoms, are asymptomatic, or are presumed to have "recovered" from past infection. The researchers hope to expand COVID-19 testing options across Canada and ultimately, better identify patients who carry high risk of community transmission than traditional respiratory testing alone.

Cellular Immuno-Therapy for COVID-19 Induced Acute Respiratory Distress Syndrome: The CIRCA-19 TrialDuncan Stewart, Principal InvestigatorOttawa Hospital Research Institute

Through a series of trials, this research will rapidly evaluate the safety and efficacy of using mesenchymal stromal/stem cells, or MSCs, to help treat patients with COVID-19 related acute respiratory distress syndrome (ARDS). Up to 25 percent of all patients admitted to hospital require admission to an intensive care unit, and as many as 40 percent develop severe difficulty breathing due to ARDS.

In total, 27 patients will undergo three sequential trials. The first trial, called the Vanguard study, is designed to quickly determine the optimal dosing strategy of MSCs derived from bone marrow to treat patients experiencing ARDS. The next two trials will use the optimal dose of cells determined by the Vanguard trial, but will administer MSCs derived from the umbilical cord, which is an abundant and readily available source.

Rapid Identification of Immunogenic and T-cell Epitopes to Enable Serologic Testing, Passive Immunotherapy, and Epitope Vaccine for COVID-19Shawn Li, Principal InvestigatorWestern University

To curb the COVID-19 outbreak caused by the SARS-CoV-2 virus, researchers are looking to solve three critical challenges as quickly as possible - detection, treatment, and vaccination. This project will address these challenges by developing a point-of-care blood test to identify infected individuals, including those without symptoms, devising strategies for the production of virus-neutralizing antibodies to treat the severely ill, and identifying viral epitopes to inform epitope-vaccine development.

The Impact of the Coronavirus Pandemic on Children with Medical Complexity Technology Dependency: A Novel Research Cohort StudyAudrey Lim, Principal InvestigatorMcMaster University

This study addresses how to effectively manage pediatric patients remotely by identifying the barriers and facilitators of virtual clinics. COVID-19 is placing strain on families of children with medical complexity, medical fragility and technology dependency. Many of these children are dependent on life sustaining technology such as tracheostomy, home mechanical ventilation, and/or enteral feeding tubes. Though accounting for less than 1 percent of all children in Ontario, this group is at increased risk of multiple and prolonged hospitalizations and poorer health outcomes. Normally, these children are seen at a hospital to address their multiple complex needs, however due to COVID-19, all in-person clinic appointments have been replaced by virtual clinics. Parental satisfaction with virtual clinic healthcare teams will also be assessed using a quality improvement tool developed for this study. This research has the potential to advance virtual medicine, beyond COVID-19.

Food Retail Environment Surveillance for Health and Economic Resiliency: FRESHER OntarioJason Gilliland, Principal InvestigatorWestern University

The Food Retail Environment Surveillance for Health & Economic Resiliency (FRESHER) project is a rapid response to the widespread closures of, and modified operating conditions for, many retail food outlets. The FRESHER project will examine the economic and social impacts of COVID-19 in Southwestern Ontario by identifying what businesses modified their operations, temporarily closed or permanently closed during the outbreak and how the outbreak has affected businesses and their employees. This study will help inform policies and programs that will maintain Ontario's food security, incentivize economic growth during the recovery period, and improve resiliency among businesses during future pandemics and emergencies.

Protective Immunity in Individuals Infected with COVID-19Ishac Nazy, Principal InvestigatorMcMaster University

The goal of this research is to determine the makeup, concentration, strength and viral properties of anti-SARS-CoV-2 antibodies to provide insights into the immune response of individuals infected with COVID-19. Working with Dr. Arnold (CONCOR-1 study on convalescent plasma therapy), this team will use samples from recovered patients to test whether antibodies exist, and if they are able to bind and neutralize the virus. This research will determine whether immunity is longstanding or if it wanes over time; and will inform researchers how immune-based treatments work to fight off the virus, including convalescent plasma or future vaccines.

Clinical Research on the Therapeutic Benefits of Annexin A5 in Severe COVID-19 PatientsClaudio Martin, Principal InvestigatorLawson Health Research Institute

There are currently no proven therapies to treat COVID-19. In the most severe cases, the disease is complicated by sepsis acute respiratory distress syndrome (ARDS), and multiorgan failure. Sepsis is a life-threatening condition caused by the body's response to an infection. While the body normally releases chemicals to fight an infection, sepsis occurs when the body's response to these chemicals is out of balance, triggering systemic inflammation that can damage multiple organs. Many critically ill COVID-19 patients develop sepsis 1-2 days before ARDS, suggesting that sepsis is a major contributor to the development of organ and respiratory failure.

This clinical trial will examine the effects of Annexin A5, in treating critically ill COVID-19 patients who develop sepsis. Annexin A5 is a human protein that has potent anti-inflammatory, anti-apoptotic (cell death prevention) and moderate anticoagulant (blood clot prevention) properties. The ultimate goal of the trial is to use Annexin A5 to treat sepsis and prevent respiratory and multi-organ failure.

Novel Coronavirus Antiviral Drug Discovery Using High-Throughput ScreeningJean-Simon Diallo, Principal InvestigatorOttawa Hospital Research Institute

Using a novel bio-sensor that detects drugs that disrupt the attachment of coronaviruses to cells, this research will test approximately 1,200 approved drugs to better understand their potential to prevent viral infection in cells and their ability to block the interaction between COVID-19 and its receptor. A second phase of this study will attempt to identify novel antivirals from a small (>220,000) molecule library.

Canada's COVID-19 Pandemic Response and Impact in Low-Income and Homeless or At-Risk for Homelessness Populations in Ottawa (Canada): A Mixed Method StudySmita Pakhale, Principal InvestigatorThe Ottawa Hospital Research Institute

Vulnerable populations face numerous social and health inequities that are exacerbated during times of crises. Lessons learned from previous public health crises suggest that inappropriate communication strategies jeopardize risk reduction for vulnerable populations. The objective of this research is to measure the impacts of COVID-19 public health emergency response efforts and communication strategies on Ottawa's low-income, homeless or at-risk for homelessness populations. The findings could help inform public health messaging strategies and pandemic approaches for vulnerable populations.

Go here to read the rest:
Ontario Announces First Phase of Research Projects to Fight COVID-19 - Government of Ontario News

Posted in New York Stem Cells | Comments Off on Ontario Announces First Phase of Research Projects to Fight COVID-19 – Government of Ontario News

New Partnership Could Eventually Lead To One-Size-Fits-All Cancer Vaccines – Forbes

Posted: January 29, 2020 at 5:43 pm

British scientists who announced last week their discovery of a new type of cancer-killing T-cell have entered a partnership with a biotechnology company pioneering the use of Dark Antigens to developT-cell receptor (TCR)-based immunotherapies and off-the-shelf cancer vaccines. The resultthey hopewill be a one-size-fits-all cancer therapy.

Last Monday, scientists at Cardiff University in the UK announced they had identified a new type of killer T-cella T-cell clonethat recognized and killed multiple different types of human cancer, while ignoring healthy, non-cancerous cells. The discovery, researchers said, offers hope of a universal cancer therapy. The researchers reported in Nature Immunology that these T-cells attacked many forms of cancer from all individuals. The T-cell clone killed lung, skin, blood, colon, breast, bone, prostate, ovarian, kidney and cervical cancer.

Less than a week later, the Cardiff researchers have announced they will enter a partnership with Ervaxx to eventually bring their discovery to patients.

Cardiff University Professor Andrew Sewell

The Cardiff University T-cell modulation group, within the Division of Infection and Immunity, studies all areas of T-cell biology including T-cell genetics, molecular biology, protein chemistry, crystallography and cell biology. The group aims to understand the genetic, biochemical and cellular mechanisms that govern T-cell responses in human diseases, such as HIV, EBV, tuberculosis autoimmunity and cancer.

Professor Andrew Sewell with Research Fellow Garry Dolton

Ervaxx is a UK biotechnology company based in London and Oxford, which is pioneering a new approach to developing targeted immunotherapies for treating and preventing cancer. These immunotherapies, including T-cell therapies, are based on new cancer targets (Dark Antigens) that derive from the dark matter of the genome, which are generally silenced in normal tissue but can become selectively activated in cancer.

T-cell therapies for cancer are the latest paradigm in cancer treatments. Current therapies include CAR-T and TCR-T, where immune cells are removed, genetically-modified and returned to a patients blood to seek and destroy cancer cells. Current therapies are personalized to each patient, target only a few types of blood cancer and have not been successful for solid tumors, which make up the vast majority of cancers.

In contrast, the newly discovered cell attaches to a molecule on cancer cells called MR1, which does not vary in humans.So not only would the treatment work for most types of cancer, said Professor Andrew Sewell, an expert in T-cells and a lead author on the study from Cardiff Universitys School of Medicine, but the same approach could be applied in all patients. It is hoped that the approach might eventually be applied as an almost instant off-the-shelf treatment.

The use of HLA-agnostic T-cell receptors has the promise to transform the treatment of common solid tumors that are presently incurable, said Carl June, MD, in reference to the Cardiff research. A leading expert in the delivery of successful T-cell therapies, June is a professor in Immunotherapy in the Department of Pathology and Laboratory Medicine and the director of the Center for Cellular Immunotherapies and of Translational Research in the Abramson Cancer Center of the University of Pennsylvania. As with organ or bone marrow transplants, previously identified cancer-specific T-cells have been suited only to small sections of the population who share specific tissue types, making it difficult to identify and treat the most appropriate patients. This new T-cell appears not to have these limitations, and if this is borne out in clinical testing, and the approach is shown to be safe and efficacious, it could represent a real advance for the field. We need to cure cancer and not turn it in to a chronic disease.

June studies various mechanisms of lymphocyte activation that relate to immune tolerance and adoptive immunotherapy for cancer and chronic infection. According to the Parker Institute, his research team published findings in 2011, which represented the first successful and sustained demonstration of the use of gene transfer therapy to treat cancer. Clinical trials utilizing this approach, in which patients are treated with genetically engineered versions of their own T-cells, are now underway for adults with chronic lymphocytic leukemia and adults and children with acute lymphoblastic leukemia. Early results in that group show that 90 percent of patients respond to the therapy, and more recently, trials of this approach have begun for patients with other blood cancers and solid tumors including pancreatic cancer, mesothelioma and the brain cancer glioblastoma. In 2017, it became the United States first FDA-approved personalized cellular therapy for the treatment of cancer.

Still, Sewell cautioned people from becoming overly optimistic too soon about Cardiffs findings. He said while the scientists discovery is potentially game-changing, an actual universal cancer therapy could be years away.I would really like to stress that we have not cured a patientour results were all laboratory based, albeit with patient T-cells and cancer cells. Clearing cancer in a culture dish and clearing it in a patient are two very different things.

When Cardiff researchers injected the new immune cells into mice with a human immune system and a human blood cancer line, the cancers cells were cleared to a level seen with CAR-T cells in the same mouse model, Sewell said. The group further demonstrated that equipping T-cells of skin cancer patients with the new receptor induced them to destroy not only the patients own cancer cells, but also other patients cancer cells in the laboratory, he said.

Cardiff researchers have now discovered T-cells equipped with a new type of T-cell receptor (TCR) which recognizes and kills most human cancer types, while ignoring healthy cells, Cardiff reported in a press release. This TCR recognizes a molecule present on the surface of a wide range of cancer cells as well as in many of the bodys normal cells but, remarkably, is able to distinguish between healthy cells and cancerous ones, killing only the latter.

Though there are various types of T-cells, Sewell said his interest is in killer T-cellsalso called cytotoxic T-cells. Killer T-cells are fascinating as they have the unique ability to see inside other body cells and scan them for anomalies he said. Conventionally, killer T-cells scan the molecular machines inside cells called proteins. A clever system presents bits of all the proteins inside each cell on its surface bound to molecular platforms called HLA [Human Leukocyte Antigen]. Normal, healthy body cells only present bits of normal proteins, and these are ignored by killer T-cells. If a cell is, for instance, infected with a virus, then it will contain some proteins of viral origin and bits of these will be displayed on the surface of the infected cell. Killer T-cells can recognize these protein fragments as foreign. This activates the killer T-cell to destroy the infected body cell and all its contents, including the virus. In this sense, killer T-cells act as a sophisticated seek and destroy weapon.

T-cells attacking cancer.

When cells become cancerous, they change the expression of some proteins and some proteins mutate, Sewell explained. These changes can also be detected by killer T-cells. Successful cancers go to great lengths to hide from killer T-cells, he said. We know that cancer often exploits the safety checkpoints that are built into T-cells to prevent them causing inflammation or autoimmunity. These checkpoints can be thought of as T-cell brakes, and successful cancers are often good at applying these brakes. Recent development of new drugs called checkpoint inhibitors prevents the application of these brakes and can result in complete clearance of some cancers in some people. Research that led to the discovery of checkpoint inhibitors was awarded the Nobel prize for Physiology or Medicine in 2018.

Sewell said the Cardiff teams discovery could mean exciting opportunities for pan-cancer, pan-population immunotherapies not previously thought possible. The research was funded by the Wellcome Trust, Health and Care Research Wales and Tenovus.

Until now, Sewell said, nobody knew this cell existed. He said the teams hypothesis is that the T-cell works by interacting with a molecule called MR1 which, in turn, flags up the distorted metabolism in a cancer cell.

Now that we know that these types of cells exist, we can actively look for others that work by a similar mechanism. Indeed, we have already found similar broadly tumoricidal, HLA-agnostic killer T-cells that see cancers via different surface molecules. The molecules targeted by these cells were also discovered using the CRISPR library approach. CRISPR gene editing has been a real game-changer.

Sewell said the next step will begin with safety testing on further healthy human cell lines in the laboratory. History has shown, he said, that some T-cells could attack things we dont want them to. We have already demonstrated that our new T-cell does not respond to 20 healthy cell types, he said. The human body has many more cell types than this so, as best we can, we need to rule out that this T-cell does not attack any further healthy human cell types. The new MR1-binding receptor has a natural sequence isolated from a healthy donor and thus the likelihood that it will attack healthy tissue is unlikely.

In any event, Sewell said it is important for people to acknowledge that this discovery has not been tested outside of the laboratory and not yet in human beings. It is impossible to reconstitute a whole human body as individual cell types in the laboratory, so after passing an accepted level of safety testing in this way, the next step is a first-in-man trial Sewell said. In order to minimize the risk, it is likely that the first time this type of T-cell is used in man, the T-cells will transiently [impermanently] express the relevant T-cell receptor and be given in low numbers, with escalation from there once safety is demonstrated. This way if there is any autoimmune attack it will be at low level and short-lived, so hopefully do minimal damage.

While Sewell hesitated at giving a timeline for when an actual universal cancer therapy or cure could be expected, he is hopeful that clinical trials may start in the next few years once further laboratory safety testing is completed.

The new collaboration to develop this recent discovery funded by Ervaxxwill support a multi-year research program with Sewells T-cell modulation group at Cardiff University focusing on the discovery and characterization of T-cells and TCRs reactive to cancer-specific antigens and ligands, including Ervaxx proprietary Dark Antigens.

The company has the right to advance resulting candidate T-cell/TCR-based immunotherapeutics and cancer vaccines through development and commercialization, Ervaxx stated in a press release.

Kevin Pojasek, Ervaxx CEO, said the collaboration with the Cardiff University research group shows early but enormous potential for the treatment of cancers. He said the partnership, which follows those with the University of Oxford, University of Cambridge and Johns Hopkins University School of Medicine, reinforces our ambition to collaborate with leading academic institutions and be at the cutting edge of the T-cell immunology field to drive the development of novel off-the-shelf cancer therapies.

In terms of the MR1 finding, when asked if it meant that some people are completely immune to cancer, Sewell said, Possibly. This immune cell could be quite rare, or it could be that lots of people have this receptor, but for some reason it is not activated. We just don't know yet, but we hope that this finding can be exploited and will pave the way for new cancer treatments.

View post:
New Partnership Could Eventually Lead To One-Size-Fits-All Cancer Vaccines - Forbes

Posted in Cell Medicine | Comments Off on New Partnership Could Eventually Lead To One-Size-Fits-All Cancer Vaccines – Forbes