Fact-checked by Grok 2 weeks ago

Moderna

Moderna, Inc. is an American company specializing in the research, development, and commercialization of (mRNA)-based therapeutics and vaccines for serious diseases, including infectious diseases, immuno-oncology, rare diseases, and cardiovascular conditions. Founded in 2010 by , Robert Langer, and through , the company is headquartered in , and has pioneered mRNA technology to instruct human cells to produce proteins that combat illness. Under CEO , who joined in 2011, Moderna advanced its platform with strategic partnerships, culminating in the rapid creation of Spikevax (mRNA-1273), its developed in collaboration with the (NIH). The vaccine received from the U.S. (FDA) on December 18, 2020, after phase 3 clinical trials showed 94.1% efficacy in preventing symptomatic in adults, with no severe cases in the vaccinated group. This breakthrough propelled Moderna to commercial success, with Spikevax generating peak sales of $18.4 billion in 2022, though revenues later declined amid reduced demand and competition. The company's mRNA approach enabled unprecedented development speed, supported by government funding and derisking mechanisms, but has sparked disputes, including a legal battle with the NIH over inventorship for stabilizing nanoparticles critical to vaccine delivery. Beyond , Moderna expanded its portfolio with FDA approval of mRESVIA, an for older adults, in 2024, and maintains a broad pipeline targeting , , and personalized cancer therapies, reflecting ongoing innovation despite post-pandemic financial pressures and safety monitoring for rare adverse events like confirmed in real-world data.

Company Overview

Founding and Early Milestones

Moderna was founded in 2010 by , founder and CEO of , in collaboration with Robert Langer, a professor of at , and , a biologist, with the aim of developing (mRNA) therapeutics as a novel drug modality. The company originated from exploratory work within Flagship's VentureLabs, initially codenamed LS18, and was renamed Moderna Therapeutics to reflect its focus on modified RNA technologies; provisional patents for mRNA applications were filed as early as July and October 2010. Stéphane Bancel joined as CEO in October 2011, bringing prior experience from roles at and to lead operations amid the nascent field of mRNA research. Early financing included a $40 million round announced in November 2012 to advance mRNA , supported by investors including , which provided foundational backing as the incubator. Key early milestones included a multiyear partnership with AstraZeneca in 2013 to develop mRNA-based therapeutics for cardiovascular and metabolic diseases, alongside a $24.6 million grant from the Defense Advanced Research Projects Agency (DARPA) awarded on October 2, 2013, to support preclinical development of mRNA-encoded antibody candidates under the ADEPT program. In 2014, Moderna opened expanded headquarters and laboratories in Cambridge, Massachusetts, and formed a collaboration with Alexion Pharmaceuticals for rare disease applications. A significant technical achievement occurred in 2015 with the administration of the first human dose of an investigational mRNA vaccine (mRNA-1440) targeting the H10N8 influenza strain, marking the transition from preclinical to clinical testing.

Leadership and Organizational Structure

Stéphane Bancel has served as Chief Executive Officer of Moderna since October 2011, overseeing the company's strategic direction, including its expansion into mRNA-based therapeutics and vaccines. Prior to Moderna, Bancel held executive roles at and , bringing expertise in diagnostics and pharmaceuticals. The executive leadership team comprises key functional heads reporting to Bancel, including Stephen Hoge, M.D., as President, responsible for operational oversight; Jamey Mock as , managing financial strategy and accounting; Jacqueline Miller, M.D., as , leading clinical development; and Jerh Collins, Ph.D., as Chief Technical Operations and Quality Officer, handling manufacturing and . Other senior roles include Tracey Franklin as Chief People and Digital Technology Officer, a position reflecting Moderna's 2025 merger of and functions to integrate AI-driven efficiencies across and digital infrastructure. Shannon Thyme Klinger serves as Chief Legal Officer and Corporate Secretary, addressing regulatory and compliance matters. Moderna maintains a , with specialized departments in , clinical operations, , and , enabling focused expertise in mRNA platform advancement while supporting in production and commercialization. The , chaired by co-founder since the company's inception, provides governance and strategic guidance, comprising ten members including Bancel, independent directors such as Sandra Horning, M.D., former at , and Elizabeth Nabel, M.D., former President of Brigham Health, alongside experts in finance and biotech like David M. Rubenstein, Co-Founder of . The board's composition emphasizes scientific, operational, and financial acumen to align with Moderna's mission in therapeutic innovation.

Core Mission and Strategic Focus

Moderna's stated mission is to deliver on the promise of mRNA science to create a new generation of transformative medicines for patients, with a focus on using its mRNA platform and supporting infrastructure to address infectious diseases and serious conditions. This entails prompting the body's cells to produce proteins that combat disease, either by eliciting immune responses for or directly treating underlying in therapeutics. The approach prioritizes diseases lacking effective treatments, such as certain cancers, rare genetic disorders, and latent viral infections, under the principle that mRNA's versatility allows broad applicability if successful in one area. Strategically, Moderna emphasizes speed in mRNA design and iteration, enabling the testing of multiple candidates within days via computational tools and scalable , which shortens traditional development timelines from years to months. As of January 2025, the company's priorities include driving revenue growth from existing products, securing up to 10 regulatory approvals over the ensuing three years across and therapeutics, and enforcing cost discipline to achieve by 2028, amid a 41% revenue decline in the first half of 2025 largely attributable to reduced demand. This has prompted a $1.5 billion cost-cutting initiative, including R&D optimization, while maintaining investments in high-potential areas like and respiratory virus combinations. The firm's pipeline reflects this dual emphasis on mission-driven innovation and pragmatic commercialization, with ongoing programs in mRNA-based individualized neoantigen therapies for cancer, prophylactic for and , and exploratory efforts in autoimmune and cardiovascular diseases, all aimed at mitigating reliance on pandemic-era successes. features, such as avoiding animal-derived components and preservatives in formulations, underscore a commitment to scalable, equitable deployment of mRNA medicines globally.

Technological Platform

Development of mRNA Technology

Moderna's development of mRNA technology originated from foundational research in chemical modifications to , aimed at reducing innate immune activation that previously limited its therapeutic potential. In 2010, the company was established by , a biologist, alongside and Robert Langer, to commercialize Rossi's work on modified mRNA conducted at Harvard's Stem Cell Institute. Rossi's team demonstrated that incorporating nucleoside analogs, such as , into synthetic mRNA enabled efficient cellular reprogramming without triggering severe inflammatory responses, as shown in experiments converting adult fibroblasts into induced pluripotent s. This modification built on prior discoveries by and , who in 2005 reported that substitution evaded detection, but Rossi's application to transient protein expression marked a shift toward scalable therapeutics. The platform's core innovation lay in treating mRNA as a programmable , where design and chemistry were optimized iteratively to enhance stability, translation efficiency, and payload specificity. Early efforts at Moderna involved of modified mRNA variants to identify those yielding higher protein expression levels , with and proving particularly effective in suppressing responses while boosting . By 2012, the company had advanced to preclinical models, demonstrating dose-dependent protein production for applications beyond stem cells, such as cytokine replacement and vaccine antigens. This modality-centric approach emphasized repeatability across targets, contrasting with traditional biologics by enabling rapid via computational prediction rather than cell-line . Subsequent refinements addressed remaining challenges like mRNA degradation and off-target effects, incorporating 5' cap analogs and poly-A tail optimizations to mimic endogenous transcripts more closely. In 2013, Moderna secured a $25 million contract to develop mRNA-based therapeutics for infectious diseases, validating the platform's viability and funding further lipid formulation integration for delivery—though core mRNA remained the foundational step. These developments positioned mRNA as a versatile template for encoding any protein of interest, with empirical data from and non-human studies confirming expression levels up to 1,000-fold higher than unmodified . Peer-reviewed validations underscored the causal link between modifications and reduced , enabling progression to human trials by the mid-2010s.

Key Innovations in Delivery and Stability

Moderna's advancements in mRNA delivery center on proprietary (LNP) formulations, which encapsulate mRNA to shield it from extracellular degradation and enable efficient intracellular delivery. These LNPs typically comprise an ionizable cationic (such as in their mRNA-1273), a helper , for fusion, and a (PEG)- for steric stabilization. The ionizable remains neutral at physiological pH to minimize but protonates in the acidic , promoting disruption and mRNA release into the . This LNP design facilitates endocytic uptake by antigen-presenting cells, with Moderna's proprietary variants demonstrating enhanced local protein expression and reduced reactogenicity compared to non-optimized formulations, as evidenced in preclinical and clinical data from their programs. Further innovations include tailored LNPs for alternative administration routes, such as pulmonary via nebulization or intratumoral injection, expanding beyond intramuscular delivery to target mucosal or tumor microenvironments. In parallel, Moderna has pioneered mRNA stability enhancements through chemical and structural modifications that mitigate rapid enzymatic degradation and innate immune activation. A is the of all uridines with (m1Ψ), which dampens recognition by Toll-like receptors (TLRs) and RIG-I-like receptors, thereby extending mRNA half-life in cells while amplifying translational output—key to achieving robust expression in vaccines like mRNA-1273. Complementary optimizations include codon usage adjustment to favor high-expression sequences, proprietary 5' untranslated regions (UTRs) that promote efficient scanning and initiation, and 3' UTRs derived from stability-conferring human genes to prolong cytoplasmic persistence. These elements, combined with a Cap 1 structure at the 5' end and an extended poly-A tail, collectively yield mRNAs with superior durability and immunogenicity profiles over unmodified counterparts.

Historical Product Development

Pre-Pandemic Research and Trials

Moderna's pre-pandemic efforts focused on pioneering mRNA-based vaccines and therapeutics, building on foundational work in lipid nanoparticle delivery systems to encapsulate and stabilize synthetic mRNA for cellular uptake. Established in 2010, the company prioritized infectious diseases, oncology, and rare genetic disorders, conducting early-phase clinical trials to assess safety, tolerability, and immunogenicity without achieving regulatory approvals prior to 2020. Research emphasized unmodified and modified mRNA constructs to mitigate innate immune activation, a persistent hurdle in earlier mRNA studies. The company's inaugural human trials began in 2015 with Phase 1 studies for an candidate, mRNA-1440, which encoded proteins from H10N8 and H7N9 strains and elicited dose-dependent responses in healthy adults, though with notable injection-site reactions and systemic symptoms. In 2016, Moderna expanded to Phase 1 evaluations of mRNA vaccines against and virus, demonstrating neutralizing production but underscoring needs for optimized dosing to balance efficacy and reactogenicity. By 2017, a Phase 1 trial for a (CMV) vaccine, mRNA-1647, targeted B and matrix proteins to induce both humoral and cellular immunity, advancing understanding of multivalent mRNA immunogenicity in preventing congenital infections. Additional Phase 1 efforts included (RSV) and vaccines, with preclinical data supporting progression but clinical results revealing variable T-cell responses. Beyond infectious diseases, Moderna pursued mRNA therapeutics for cancer and rare diseases through strategic partnerships that facilitated trial initiation. In , a with Merck launched 1 trials of personalized neoantigen-targeted mRNA-4157 for and other solid tumors, combining mRNA with inhibitors to enhance tumor-specific T-cell responses. Agreements with Alexion (2015) for and , and Vertex (2015) for , yielded 1/2 studies of mRNA-encoded enzymes, though efficacy remained limited by transient protein expression. Funding from ($25 million in 2013 for trauma applications) and the Gates Foundation ( investment) supported platform maturation, yet trials consistently encountered delivery inefficiencies and off-target immune effects, stalling advancement to later phases.

COVID-19 Vaccine Acceleration (mRNA-1273)

In response to the January 10, 2020, public release of the genetic sequence by Chinese researchers, Moderna and the National Institute of Allergy and Infectious Diseases (NIAID) collaboratively designed the mRNA-1273 vaccine candidate within two days, encoding the full-length stabilized in its prefusion conformation. The first vial of mRNA-1273 was produced on January 13, 2020, followed by shipment to the NIH for animal studies on January 24, with confirmed in mice by early February. This rapid initiation leveraged Moderna's pre-existing mRNA , developed over a decade through prior investments in lipid delivery and mRNA stabilization technologies, which had been tested in non-COVID applications like and Zika vaccines. An application was submitted to the FDA in late February 2020, enabling the Phase 1 to commence on March 16, 2020, in healthy adults aged 18-55, evaluating safety, reactogenicity, and across escalating doses of 25-100 μg administered in two intramuscular injections 28 days apart. The accelerated timeline incorporated overlapping clinical phases and at-risk manufacturing, supported by substantial U.S. government funding through the Biomedical Advanced Research and Development Authority (BARDA). Initial BARDA awards totaling approximately $955 million facilitated preclinical scaling and Phase 1/2 trials, while an , , agreement added up to $1.525 billion to produce and deliver 100 million doses contingent on Phase 3 success, bringing total Warp Speed investment to about $2.5 billion. Phase 2 expansion in May 2020 assessed in broader age groups, including older adults and those with comorbidities, showing robust neutralizing antibody responses comparable to Phase 1. The pivotal Phase 3 COVE trial, a randomized, placebo-controlled study enrolling 30,420 participants starting July 27, 2020, evaluated efficacy against virologically confirmed occurring at least 14 days after the second dose, with primary endpoints focusing on symptomatic disease prevention. This compression of traditional sequential development—typically spanning 10-15 years—relied on prior mRNA data, sharing with regulators, and parallel process validation for Good Manufacturing Practice production. Interim Phase 3 results, analyzed by an independent data and safety monitoring board in November 2020, demonstrated 94.1% efficacy (95% CI, 89.3-96.8) in preventing COVID-19 illness among 185 confirmed cases (11 in vaccine group vs. 185 in placebo), including 100% efficacy against severe disease, based on 30 cases (zero vaccine-related). Safety data indicated mostly transient reactogenicity, with grade 3 local reactions in 2.7% after dose 1 and 16.5% after dose 2, and systemic events like fatigue (9.7%) and headache (4.5%) more common post-dose 2, but no vaccine-associated enhanced disease or excess serious adverse events beyond placebo rates. These findings prompted the FDA to issue Emergency Use Authorization for mRNA-1273 on December 18, 2020, for individuals 18 years and older, following review of manufacturing data and a 2:1 benefit-risk assessment amid surging pandemic cases. Full dataset confirmation in December 2020 upheld the efficacy and safety profile, with longer-term follow-up through May 2021 showing sustained protection beyond five months. The program's success highlighted causal factors like massive parallel funding and regulatory emergency provisions, though it involved financial risks from pre-emptive scale-up of unproven candidates.

Post-2020 Expansions and Approvals

Following the of its (mRNA-1273, branded Spikevax) on December 18, 2020, Moderna pursued label expansions and full approvals. On October 21, 2021, the U.S. FDA authorized a of Spikevax for adults aged 65 and older, as well as certain high-risk individuals aged 18-64, based on clinical data demonstrating sustained . This was followed by full Biologics License Application approval on January 31, 2022, for individuals 18 years and older, supported by phase 3 results showing 93% efficacy against symptomatic and a safety profile consistent with EUA data from over 30,000 participants. Subsequent expansions included EUAs for pediatric use in June 2022 (ages 6 months to 17 years) and bivalent boosters targeting variants in August 2022, reflecting iterative adaptations to emerging strains. Moderna continued annual updates to Spikevax formulations, with the FDA approving the 2024-2025 version on August 22, 2024, for ages 12 and older, incorporating antigens matched to circulating variants like KP.2. In July 2025, full approval was extended to high-risk individuals aged 6 months through 64 years. A next-generation , mNEXSPIKE (mRNA-1283), received FDA approval in May 2025 for adults, demonstrating superior neutralizing antibody responses compared to Spikevax in phase 3 trials, marking Moderna's first approved mRNA-based update beyond the original platform. Further refinements included approvals for 2025-2026 formulas targeting the LP.8.1 variant on August 27, 2025, and September 8, 2025, respectively, for Spikevax and mNEXSPIKE in eligible populations. Beyond , Moderna's first non-COVID approval came with mRESVIA (mRNA-1345), an vaccine granted full FDA approval on May 31, 2024, for preventing lower respiratory tract disease in adults aged 60 and older, based on the ConquerRSV phase 3 showing 83.7% against RSV-associated illness over 18 months. This represented a significant pipeline expansion into respiratory viruses, leveraging mRNA technology for rapid design. On June 12, 2025, the approval was broadened to adults aged 18-59 at increased risk for severe RSV, supported by immunogenicity bridging studies and safety data from over 37,000 participants across trials. These approvals diversified Moderna's portfolio from pandemic-focused products to routine preventive vaccines, though ongoing programs in and combination vaccines remained in clinical stages without post-2020 approvals as of October 2025.

Current Products and Pipeline

Approved Vaccines and Therapeutics

Moderna has two commercially available mRNA vaccines approved by regulatory authorities: Spikevax for prevention and mRESVIA for (RSV) lower respiratory tract disease prevention. A next-generation , mNEXSPIKE, also received U.S. (FDA) approval in 2025. As of October 2025, Moderna has no approved therapeutics, with its pipeline focused on investigational treatments for , rare diseases, and infectious diseases remaining in clinical stages. Spikevax (mRNA-1273), Moderna's flagship , encodes a stabilized prefusion of to elicit immune responses. The FDA granted (EUA) on December 18, 2020, for individuals 18 years and older, followed by full approval on January 31, 2022, for the same population to prevent COVID-19. The () issued conditional marketing authorization on January 6, 2021, converted to standard authorization later. Indications expanded to include children aged 6 months through 11 years at high risk for severe disease, with full FDA approval for this group on July 10, 2025. Annual updates target circulating variants; the 2025-2026 formulation, monovalent against the LP.8.1 lineage, was FDA-approved on August 27, 2025, revoking prior EUAs for superseded versions. mRESVIA (mRNA-1345) targets the F glycoprotein in prefusion conformation to prevent -associated lower disease. The FDA approved it on May 31, 2024, for adults aged 60 years and older, based on phase 3 trial data showing 83.7% efficacy against -confirmed lower disease over 3.7 months median follow-up. authorization followed on August 15, 2024. In June 2025, the FDA expanded approval to adults aged 18 to 59 at increased risk for disease, addressing gaps in younger high-risk populations such as those with chronic conditions. mNEXSPIKE, a second-generation , was FDA-approved on September 25, 2025, for against in individuals aged 12 years and older, featuring an optimized design for potentially broader and more durable immunity compared to Spikevax. It targets variants including LP.8.1, aligning with seasonal update strategies.
ProductIndicationKey ApprovalsTarget Population
Spikevax (mRNA-1273)Prevention of FDA full approval Jan 31, 2022 (adults 18+); updates through 2025-20266 months+ (high-risk children); 12+ (broader)
mRESVIA (mRNA-1345)Prevention of RSV lower respiratory tract diseaseFDA approval May 31, 2024 (60+); expanded Jun 2025 (18-59 at risk)Adults 18+ at risk; primarily 60+
mNEXSPIKEPrevention of FDA approval Sep 25, 202512+ years

Ongoing Clinical Programs

Moderna's ongoing clinical programs primarily utilize its mRNA platform to develop vaccines and therapeutics across infectious diseases, , and rare genetic disorders, with a strategic emphasis on respiratory viruses, latent infections, cancer immunotherapies, and . Following the October 2025 discontinuation of its cytomegalovirus (CMV) vaccine candidate mRNA-1647 after it demonstrated insufficient protective efficacy in a Phase 3 against congenital CMV in women of childbearing age, the company has refocused resources on higher-priority assets. This shift aligns with broader portfolio prioritization announced in early 2025, aiming for advancements in areas with demonstrated proof-of-concept data. In respiratory vaccines, several candidates remain in late-stage development. mRNA-1010, a seasonal , is in Phase 3 trials evaluating and against contemporary strains. mRNA-1083, a combination targeting both and , is also advancing in Phase 3, with data supporting potential non-inferiority to monovalent counterparts in adults aged 50 and older. Earlier-stage efforts include mRNA-1365 for () and (hMPV) in Phase 1, building on Moderna's approved mRNA-1345, which is under further pediatric evaluation in Phase 2. Oncology programs represent a core focus, leveraging individualized neoantigen-specific immunotherapies. mRNA-4157 (intisemeran autogene), developed in collaboration with Merck, is in multiple Phase 3 trials as an adjuvant therapy, including for resected melanoma (KEYNOTE-942 follow-on), non-small cell lung cancer (NSCLC) post-resection, and NSCLC without pathologic complete response after neoadjuvant therapy; interim data from melanoma trials showed a 49% reduction in recurrence or death risk when combined with pembrolizumab. Phase 2 evaluations of mRNA-4157 extend to renal cell carcinoma, muscle-invasive urothelial carcinoma, non-muscle invasive bladder cancer, and metastatic melanoma, while Phase 1 studies explore broader solid tumors. Complementing this, mRNA-4359, a checkpoint immunotherapy targeting multiple tumor-associated antigens, is in Phase 2 for advanced solid tumors, with early signals of immune activation reported at ESMO 2025. mRNA-4106 remains in Phase 1 for solid tumors. For latent and other viral vaccines, active trials include mRNA-1608 (HSV-2 therapeutic ) in Phase 2 for healthy adults aged 18-55, assessing safety and ; mRNA-1189 and mRNA-1195 (Epstein-Barr virus vaccines) in Phase 2 for preventing and long-term sequelae, respectively; and mRNA-1468 (varicella-zoster virus) in Phase 2. programs feature mRNA-1403 in Phase 3 and mRNA-1405 in Phase 2, while candidates mRNA-1975 and mRNA-1982 are both in Phase 2. Earlier phases cover (mRNA-1644, Phase 1), Nipah (mRNA-1215, Phase 1), and (mRNA-1769, Phase 1). Rare disease therapeutics target metabolic disorders via mRNA-encoded enzymes. for is in Phase 2, evaluating liver expression of propionyl-CoA carboxylase in pediatric and adult patients. Similarly, for is in Phase 2, focusing on replacement. Phase 1 programs include for type 1a and a collaboration with on for , assessing alternative CFTR mRNA delivery. These efforts aim to provide transient, repeat-dosable protein replacement without genomic integration risks associated with gene therapies.

Recent Developments and Setbacks

In 2025, Moderna reported positive topline results from a Phase 3 trial of its seasonal candidate mRNA-1010, demonstrating a relative efficacy of 26.6% (95% CI: 16.7%-35.4%) against compared to a licensed standard-dose quadrivalent in adults aged 50 and older. The company plans to submit regulatory filings for approval based on these data, which also included endpoints meeting superiority criteria. Additionally, Moderna presented data on mRNA-1010 and an H5 candidate at IDWeek 2025, highlighting ongoing advancements in respiratory vaccines. The company's vaccine mRESVIA (mRNA-1345) received FDA approval on May 31, 2024, for adults aged 60 and older to prevent lower disease caused by RSV, with expanded approval on June 13, 2025, for high-risk adults aged 18 to 59. Preliminary Phase 4 results in September 2025 showed the updated COVID-19 vaccine formula eliciting an eight-fold increase in neutralizing antibodies against the LP.8.1 variant. However, Moderna discontinued development of its (CMV) vaccine mRNA-1647 on October 23, 2025, after a Phase 3 failed to meet its primary efficacy endpoint of preventing primary CMV infection in women aged 16 to 40, with the vaccine showing limited protective effect against congenital CMV transmission. Previously viewed as a potential multibillion-dollar product, the halt aligns with broader cost-cutting measures, including a $4 billion R&D budget reduction announced in September 2024. In January 2025, Moderna slashed its sales guidance for 2024 and 2025, citing lower-than-expected uptake, leading to a 17% drop. Despite these challenges, the company continues to prioritize its and pipeline, with ongoing Phase 3 programs in cancer vaccines.

Financial Trajectory

Revenue Generation and Sources

Moderna generates primarily through commercial sales of its approved mRNA vaccines, with product sales comprising approximately 97% of in recent years. The company's , Spikevax (also known as mRNA-1273), a , has been the dominant source, contributing $3.1 billion in net sales for fiscal year 2024, down from higher pandemic-era volumes due to reduced global demand for boosters. In the second quarter of 2025, Spikevax sales totaled $114 million, including $88 million from the U.S. market. A secondary product line emerged in 2024 with the U.S. FDA approval of mRESVIA (mRNA-1345), an vaccine for adults aged 60 and older, which generated $15 million in sales during the fourth quarter of 2024 as initial ization began. This launch marked Moderna's transition to a multi-product company, though RSV sales remain nascent and are projected to grow amid seasonal demand and potential expansions to other age groups or regions. Overall, fiscal 2024 reached $3.236 billion, reflecting a 52.75% year-over-year decline from 2023's $6.848 billion, driven by the expiration of advance purchase agreements and shifts to commercial markets. Grant , accounting for about 1.3% of totals, stems from government and philanthropic funding for , including historical support from entities like the U.S. Biomedical Advanced Research and Development Authority (BARDA) for early mRNA platforms. Collaboration and licensing , roughly 1.2%, arises from partnerships such as the co-development agreement with Merck for an individualized neoantigen therapy (mRNA-4157/V940) targeting , which provides milestone payments but minimal ongoing contributions absent commercial breakthroughs. For 2025, Moderna forecasts total of $1.5 billion to $2.5 billion, anticipating persistent sales pressure offset partially by RSV uptake, with no significant new streams from pipeline candidates until further approvals.

Profit Margins and Cost Structures

Moderna's gross profit margins peaked during the COVID-19 pandemic due to the scalability of its mRNA technology, which minimized variable production costs relative to high government-contracted pricing for Spikevax (mRNA-1273). In 2020, the gross margin reached approximately 96%, reflecting near-zero marginal costs after initial development, as mRNA synthesis avoids complex biological manufacturing processes typical of traditional vaccines. This declined to 85% in 2021 and 71% in 2022 amid expanding production facilities and inventory write-downs, then fell sharply to 30% in 2023 as vaccine demand waned and cost of sales rose to represent a larger revenue share. By 2024, gross margins recovered modestly to around 52-55%, supported by cost reductions in manufacturing, though still pressured by excess capacity and lower volumes.
YearGross Margin (%)
202096.03
202185.19
202270.62
202329.65
202452.50
Operating and net margins have been negative in non-pandemic years, driven by substantial fixed costs. Operating margins stood at -106% in recent as of mid-2025, with net margins at -94%, reflecting persistent operating losses despite gross profitability. Cost of sales, which includes raw materials, fill-finish operations, and royalties, dropped 69% to under $1.5 billion in 2024 from 2023 levels, comprising a higher percentage of shrinking revenues due to underutilized facilities built for pandemic-scale output. The company's cost structure remains R&D-intensive, characteristic of a clinical-stage biotech transitioning to commercialization. Research and development expenses totaled $4.84 billion in 2023, falling to $4.54 billion in 2024 amid program prioritization, representing over 60% of operating expenses. Selling, general, and administrative costs were $1.55 billion in 2023, reduced to $1.17 billion in 2024 through efficiency measures. Total operating expenses declined 35% year-over-year to $7.18 billion in 2024, with further cuts planned, including a 10% workforce reduction by end-2025 and R&D targeting $3.4 billion by 2027, to address revenue declines from $6.8 billion in 2023 to projected $1.5-2.5 billion in 2025. This structure underscores vulnerability to demand fluctuations, as high upfront investments in mRNA platform expansion—over $1 billion annually in capital expenditures pre-2023—yield low variable costs but expose the firm to losses when product sales falter.

Stock Performance and Investor Relations

Moderna, Inc. (NASDAQ: MRNA) completed its initial public offering on December 6, 2018, pricing shares at $23 and raising approximately $604 million. The stock traded modestly in its early years, reflecting the company's pre-revenue status focused on mRNA research, with a 28.09% gain in 2019. The advent of the COVID-19 pandemic catalyzed explosive growth, as Moderna's mRNA-1273 vaccine candidate advanced rapidly under Operation Warp Speed. Shares surged 434.1% in 2020 and an additional 143.11% in 2021, peaking at $497.49 on August 10, 2021, driven by emergency use authorization, widespread adoption, and peak revenues exceeding $18 billion in 2021. Post-2021, performance reversed sharply amid declining vaccine demand, competition from updated formulations, and broader market skepticism toward biotech valuations without diversified revenue. Annual returns were -29.28% in 2022, -44.63% in 2023, -58.19% in 2024, and -35.69% year-to-date through October 2025.
YearAnnual % Change
2019+28.09%
2020+434.1%
2021+143.11%
2022-29.28%
2023-44.63%
2024-58.19%
2025 (YTD)-35.69%
As of October 24, 2025, MRNA closed at $26.78, with a of approximately $10.25 billion, representing a stark contraction from pandemic-era highs exceeding $200 billion. This trajectory underscores vulnerability to single-product reliance, with trailing twelve-month falling to $3.08 billion as of June 30, 2025, primarily from boosters amid reduced global uptake. Moderna's investor relations efforts center on transparency via its dedicated website, featuring filings, quarterly earnings releases, and updates. The company hosts regular events, including earnings calls, R&D days, and analyst presentations—such as the 2024 R&D Day outlining development strategies—and maintains alerts for shareholders. Recent communications emphasize mRNA platform diversification into , flu, and , though investor focus remains on execution risks and cash burn exceeding $4 billion annually in recent quarters. No specific IR leadership details are prominently disclosed, with inquiries directed through general channels.

Controversies and Debates

Safety Profile and Adverse Events

The , Spikevax (mRNA-1273), demonstrated a profile in 3 trials characterized primarily by mild to moderate reactogenic events, including injection-site pain (92% after dose 1, 91% after dose 2), (70% and 80%), (65% and 69%), (62% and 82%), and (45% and 71%), with systemic reactions more frequent after the second dose among adults aged 18-65. Severe adverse events were uncommon during trials, occurring in approximately 0.6% of recipients versus 0.5% in , with no significant imbalance attributed to the vaccine beyond expected reactogenicity. Post-approval surveillance through systems like VAERS and V-safe has confirmed these patterns in billions of administered doses globally, though passive reporting systems like VAERS detect signals requiring further verification rather than establishing causality. Rare but serious adverse events include and , with the highest observed risk following the second dose in males aged 12-24 years, at rates of approximately 40-70 cases per million doses in this demographic according to FDA analyses. The FDA updated labeling on June 25, 2025, to reflect this risk, noting incidence peaks within 7 days post-vaccination and higher rates with mRNA-1273 compared to other mRNA vaccines in young adults. CDC data indicate overall rates post-vaccination remain rare (about 1-10 per 100,000 doses across populations) and lower than hospitalization risks from infection itself, though absolute risks prompted expanded cardiac monitoring recommendations for adolescents. occurs at rates of 2-5 per million doses, predominantly in individuals with prior allergies, leading to updates. Long-term post-approval studies, including cohort analyses up to 2-3 years post-vaccination, have not identified elevated risks for neurological disorders, autoimmune conditions, or beyond trial findings, with and sustained against variants. For the 2024-2025 updated formula targeting XBB.1.5 and JN.1 lineages, self-controlled case series data showed no increased signal for relative to prior versions, maintaining a comparable profile in over 10 million monitored doses. Multi-site global evaluations of adverse events of special interest (AESI) confirmed elevated relative risks for / (observed-to-expected ratios of 3-6 in young males) but no broad signals for , Guillain-Barré syndrome, or beyond background rates. These findings derive from regulatory and peer-reviewed observational data, though limitations in underreporting and by infection status persist in real-world assessments.

Efficacy Claims Versus Empirical Outcomes

The phase 3 of Moderna's mRNA-1273 , conducted from July 27 to , 2020, among 30,420 participants, demonstrated 94.1% (95% [CI], 89.3 to 96.8%) in preventing confirmed symptomatic illness, with the primary endpoint measured at least 14 days after the second dose. against severe was 100%, as no severe cases occurred in the vaccinated group (n=185 cases total prevented), compared to 30 severe cases in the placebo group. These results, derived under controlled conditions against early strains, formed the basis for by the FDA on December 18, 2020. Initial real-world studies aligned closely with trial data for the original strain and Alpha variant, reporting 90% effectiveness (95% CI, 84 to 94%) against infections (symptomatic or asymptomatic) among fully vaccinated U.S. healthcare personnel evaluated from December 14, 2020, to March 13, 2021. Similarly, a found 93% effectiveness against hospitalization within 91 days post-second dose. However, these estimates diminished with time and variant emergence; vaccine effectiveness against infection waned to approximately 45% by 5-6 months post-second dose against , and further to 21% (95% CI, 7-34%) against Omicron BA.1 for primary series recipients. Against Omicron symptomatic infection, effectiveness fell below 20% at six months, though boosters restored it to 61% short-term. Empirical data highlighted discrepancies in transmission prevention, with vaccines showing limited impact on Omicron infection rates despite trial focus on symptomatic disease reduction. Protection against severe outcomes persisted longer, with 95% against Omicron hospitalization after a third dose, but breakthrough infections remained common, occurring at rates up to 0.66 per 1,000 person-days post-vaccination in high-exposure groups. Hospitalization risk post-breakthrough was 40% lower and mortality 75% lower among vaccinated versus unvaccinated individuals, though against inpatient outcomes waned over time, dropping from initial highs to require booster reinforcement. These patterns, observed in diverse cohorts including older adults, underscored that trial against a static ancestral strain overestimated durability against mutating variants and prolonged community exposure. Moderna has been embroiled in multiple lawsuits related to its mRNA technology and , Spikevax, reflecting the competitive landscape of lipid nanoparticle (LNP) delivery systems and mRNA stabilization methods that underpin the platform. These disputes often stem from foundational innovations in mRNA therapeutics, where Moderna asserts on key patents developed over a decade, while challengers claim or independent invention. Outcomes have varied across jurisdictions, with some courts upholding Moderna's claims and others invalidating patents or favoring defendants, highlighting the technical complexities of proving infringement in biotech. A prominent conflict involves and , whom Moderna accused of infringing three U.S. patents covering mRNA modifications and LNP formulations in their Comirnaty . Filed in the U.S. District Court for the District of on August 26, 2022, the suit alleged that Pfizer/BioNTech reverse-engineered Moderna's technology after public disclosures, leading to billions in sales. Moderna secured a preliminary pause in April 2024 pending Patent Trial and Appeal Board (PTAB) review, but the PTAB invalidated two of the patents in March 2025, prompting Pfizer's countersuit claiming all three were invalid due to obviousness over prior publications. Internationally, Moderna prevailed in a court in March 2025, which found infringement and awarded damages, while a Court of Appeal upheld one patent's validity against Pfizer/BioNTech in August 2025 but invalidated another. Disputes with the (NIH) center on inventorship of a key for the vaccine's stabilizing . In December 2020, Moderna filed a excluding three NIH scientists who collaborated on the design under a 2013-2015 agreement, prompting NIH demands for co-inventor status in 2021. The conflict escalated to interference proceedings, with NIH arguing joint conception based on shared lab data and emails. In February 2023, Moderna paid $400 million as a "catch-up" fee for NIH's lipid-modifying technique but maintained the inventorship battle, which remains unresolved and could affect shares if NIH prevails, given the vaccine's $30+ billion in U.S. sales. Arbutus Biopharma and Genevant Sciences have sued over LNP patents essential for mRNA delivery, claiming infringement in Spikevax's encapsulation method. The initial U.S. suit in February 2022 in the District of alleged violation of patents dating to 2009-2012, with Moderna countersuing for invalidity and non-infringement based on independent LNP development. By March 2025, plaintiffs expanded to five international actions in and , seeking injunctions and damages for products beyond Spikevax. A May 2025 court ruling partially favored Arbutus on one patent, but U.S. testing disputes—where Moderna challenged Arbutus's infringement assays as flawed—continue to delay resolution, underscoring LNP's role as a in mRNA scalability. Other actions include ' suits over RNAi-enhanced mRNA tech, settled confidentially in September 2025 after Moderna fended off a U.S. claim in October 2024; GlaxoSmithKline's October 2024 Delaware suit alleging infringement in COVID and candidates; and Northwestern University's October 2024 claim on antibody-derived stabilization. These cases illustrate systemic challenges in mRNA , where overlapping claims from and early biotech contributors test enforceability amid rapid commercialization.

Ethical and Regulatory Scrutiny

Moderna's received (EUA) from the U.S. (FDA) on December 18, 2020, following a review process that prioritized rapid deployment amid the , though critics argued the expedited timeline compromised traditional standards for long-term data. The EUA required evidence supporting, rather than definitively proving, and efficacy, enabling faster rollout but drawing scrutiny for potentially lowering evidentiary thresholds compared to full approval. Regulatory concerns intensified over perceived conflicts of interest, including a "" between FDA personnel and Moderna, where former agency officials joined the company, raising questions about impartial oversight in approvals and post-market surveillance. In 2023, FDA advisers expressed disappointment and anger at Moderna and scientists for withholding early bivalent booster , relying instead on press releases rather than full peer-reviewed submissions, which undermined transparency in decision-making. Ethically, Moderna's development benefited from substantial U.S. government funding, including over $1 billion from Operation Warp Speed and National Institutes of Health (NIH) contributions to key stabilizing lipids in the mRNA formulation, yet the company disputed NIH co-inventorship on core patents, refusing to share rights or revenues despite public investment. This stance was criticized as violating the implicit social contract for taxpayer-funded research, prioritizing proprietary control over equitable global access and returns on public investment. Patent disputes extended to litigation with competitors like Pfizer-BioNTech, where Moderna's claims on mRNA delivery technologies were partially invalidated by the Patent Trial and Appeal Board in 2025, highlighting aggressive IP strategies amid ethical debates on innovation versus monopolistic barriers to derivatives. Clinical trial ethics drew attention for accelerated protocols that skipped certain preclinical steps and involved unblinding placebo groups post-efficacy demonstration, obligating vaccine access but potentially biasing long-term comparative data. Broader mRNA platform scrutiny emerged in when the U.S. Department of Health and Human Services canceled a $590 million for Moderna's H5N1 bird flu candidate, citing scientific and ethical deficiencies in efficacy data against upper respiratory infections. Regulatory bodies like the UK's Prescription Medicines Authority sanctioned Moderna multiple times, including in 2024 and , for promotional breaches and unacceptable lapses in recruitment and claims, eroding industry confidence.

Broader Impact

Scientific and Medical Contributions

Moderna's primary scientific contribution lies in advancing (mRNA) as a platform for and therapeutics, enabling rapid design and production of biologics that instruct cells to produce specific proteins for or treatment. The company's lipid nanoparticle delivery systems and mRNA modifications, such as alterations to reduce , addressed longstanding challenges in mRNA and , facilitating clinical of the . The mRNA-1273 vaccine (Spikevax), developed in collaboration with the National Institute of Allergy and Infectious Diseases, represented the first approved mRNA-based prophylactic vaccine, targeting the spike protein. Phase 3 trials demonstrated 94.1% efficacy against symptomatic disease 14 days after the second dose in adults, with sustained protection against hospitalization from early variants including alpha, , and . The vaccine's design leveraged sequence data released in January 2020, achieving by the U.S. Food and Drug Administration (FDA) on December 18, 2020, and full approval for individuals 18 years and older on January 31, 2022, marking a in vaccine manufacturing speed and scalability. Beyond , Moderna's mRNA-1345 (mRESVIA) vaccine for () received FDA approval on May 31, 2024, for adults aged 60 and older, with expansion to adults 18-59 at increased risk on June 13, 2025. Clinical data from trials involving over 35,000 participants showed 83.7% against lower disease in the first season post-vaccination, persisting at 48.3% through a second RSV season, and an 84% reduction in risk four months post-dose. Moderna's extends mRNA applications to immuno-oncology, rare diseases, and additional infectious agents, including individualized neoantigen cancer like mRNA-4157 (in with Merck), which targets tumor-specific mutations for and other cancers, and candidates for , , and . These efforts underscore mRNA's versatility for , with ongoing phase 3 trials evaluating efficacy in preventing disease progression in rare genetic disorders and eliciting T-cell responses against solid tumors.

Economic and Policy Influences

Moderna's development and deployment of its mRNA-1273 were substantially enabled by U.S. government economic interventions under , which provided approximately $2.5 billion in funding, including nearly $1 billion for and $1.5 billion for manufacturing and procurement of 100 million doses. This financial support, drawn from congressional appropriations like the , mitigated the high risks of biotech innovation by guaranteeing purchase commitments and covering production scale-up costs, allowing Moderna—a with no prior commercial products—to transition from preclinical stages to global distribution within a year. Absent such subsidies, Moderna's path to market would likely have faced prolonged private funding constraints, as evidenced by its pre-2020 reliance on and partnerships without revenue generation. The vaccine's rollout contributed to broader economic recovery by averting severe healthcare expenditures and productivity losses from ; analyses indicate that updated iterations prevented hospitalizations and yielded returns of $1.10 to $2.60 per dollar invested through reduced medical costs and sustained participation. In jurisdictions with high rates, including Moderna's product, GDP rebounds were facilitated by lowered infection-driven and hospital overloads, with collectively credited for enabling policy shifts toward reopening economies and reducing fiscal stimulus needs. However, these benefits were uneven, as global access disparities—exacerbated by Moderna's initial focus on high-income markets via advance purchase agreements—limited equitable economic gains in low-resource settings. On the policy front, expedited regulatory frameworks, such as the FDA's granted on December 18, 2020, were pivotal in accelerating Moderna's approval, bypassing traditional multi-year timelines while relying on interim trial data. This approach, rooted in declarations, set precedents for future biotech approvals but drew criticism for potential conflicts, including a documented "" between FDA officials and Moderna personnel. Moderna's success, in turn, influenced subsequent policies by validating mRNA platforms for rapid response, informing strategies like booster authorizations and combination vaccines against variants and . Yet, by 2025, policy headwinds emerged, with the administration canceling approximately $500 million in mRNA and GOP-led initiatives scrutinizing the technology amid debates, reflecting a shift from wartime acceleration to reevaluation of long-term commitments. These dynamics underscore how initial policy favoritism propelled Moderna's ascent, while empirical post-rollout data fueled countervailing regulatory caution.

Public and Expert Reception

Public reception of Moderna initially centered on acclaim for its rapid development of the mRNA-1273 (Spikevax) under , which contributed to widespread vaccination campaigns and was credited with reducing severe outcomes during the pandemic's peak. However, by 2024-2025, polls indicated significant erosion in trust, with 60% of Americans reporting they would probably not receive an updated 2024-25 , reflecting broader hesitancy toward boosters from manufacturers like Moderna. Similarly, a KFF survey in August 2025 found 59% of the public unlikely to get a that fall, amid concerns over necessity, side effects, and evolving variant data. Skepticism intensified due to perceptions of overstated long-term against and , as real-world studies showed waning protection requiring repeated boosters, alongside reports of rare but serious adverse events like , particularly in young males after the second dose. Public discourse also highlighted pricing controversies, with Moderna planning post-pandemic doses at around $130 each—far above government-purchased rates—drawing criticism for profiting from publicly funded . This fueled debates over corporate accountability, as evidenced by congressional scrutiny and analyses questioning the balance between public investment and private gains. Expert reception has been more divided than uniformly positive, with mainstream health authorities like the CDC affirming the vaccine's 89% against hospitalization in trials and a favorable overall benefit-risk profile in post-marketing data. Infectious disease specialists have defended mRNA technology's and role in saving millions of lives, citing decades of foundational and low rates of severe events. However, some analyses noted limitations, including higher systemic reactions post-second dose and challenges with variant escape, prompting calls for transparent, independent long-term surveillance beyond manufacturer-led studies. Critics, including bioethicists, have raised concerns about disputes and the platform's novelty introducing unforeseen risks, though peer-reviewed leans toward endorsement for high-risk groups while acknowledging public trust erosion from perceived regulatory shortcuts.

References

  1. [1]
    Our Mission & Values - Moderna
    Moderna's mission is to deliver the greatest possible impact to people through mRNA medicines, focusing on infectious and serious diseases.Missing: founders | Show results with:founders
  2. [2]
    Meet the Moderna Team
    Meet our board of directors ; Noubar Afeyan, Ph.D. · Co-founder and Chairman, Moderna; CEO, Flagship Pioneering ; Stéphane Bancel. Chief Executive Officer.
  3. [3]
    With Flagship Behind It, Moderna Quickly Scaled From Startup To ...
    Nov 16, 2020 · Cambridge, Massachusetts-based Moderna was founded with a mission around discovery and development of messenger RNA (mRNA) therapeutics and ...
  4. [4]
    Leadership Team - Moderna Investor Relations
    Moderna's executive team includes CEO Stéphane Bancel, President Stephen Hoge, and CFO Jamey Mock. Noubar Afeyan is Co-founder and Chairman.
  5. [5]
    [PDF] Package Insert - SPIKEVAX - FDA
    Jul 3, 2025 · 6.1 Clinical Trials Experience​​ Upon issuance of the Emergency Use Authorization (December 18, 2020) for Moderna COVID-19 Vaccine (SPIKEVAX), ...
  6. [6]
    Efficacy and Safety of the mRNA-1273 SARS-CoV-2 Vaccine
    Dec 30, 2020 · The mRNA-1273 vaccine showed 94.1% efficacy at preventing Covid-19 illness, including severe disease. Aside from transient local and systemic reactions, no ...
  7. [7]
    How Moderna's people strategy helped create its COVID vaccine
    Nov 5, 2021 · Moderna, founded in 2010, has long been a company that thrived at a ... ” Ultimately, they used their stated mission and their core ...<|separator|>
  8. [8]
    The NIH-Moderna Vaccine: Public Science, Private Profit, and ...
    As Sarpatwari shows, Moderna received unprecedented “derisking” in the form of research funding, support for clinical trials, and an advanced market commitment; ...
  9. [9]
    What the Moderna–NIH COVID vaccine patent fight means ... - Nature
    Nov 30, 2021 · Collaborators are locked in a high-stakes dispute over which researchers should be named as inventors on a key vaccine patent application.Missing: controversies empirical
  10. [10]
    [PDF] Moderna 2024 Annual Report
    Feb 21, 2025 · Despite the challenges in 2024, we became a multi-product company with the approval of mRESVIA®, our RSV vaccine for older adults.
  11. [11]
    FDA Approves Required Updated Warning in Labeling of mRNA ...
    Jun 25, 2025 · In a post-approval U.S. study funded and co-authored by FDA and published in September 2024, follow-up information was collected on ...
  12. [12]
  13. [13]
    Moderna - Flagship Pioneering
    Stéphane Bancel who had been Chief Executive Officer of the French diagnostics company bioMérieux SA, joined Moderna's board of directors in March 2011, and ...
  14. [14]
    Moderna Announces $40 Million In Financing To Advance ...
    Board members Robert Langer and Derrick Rossi are academic co-founders of Moderna, along with scientific advisory board member Kenneth Chien from Harvard ...
  15. [15]
    Our Story - Moderna
    Since our founding in 2010, we have worked to build the industry's leading mRNA technology platform.
  16. [16]
    DARPA Awards Moderna Therapeutics a Grant for up to $25 Million ...
    Oct 2, 2013 · This $24.6 million grant could support research for up to 5 years to advance promising antibody- producing drug candidates into preclinical ...Missing: milestones | Show results with:milestones
  17. [17]
    Stéphane Bancel - Forbes
    He became CEO of Moderna in 2011 and owns a roughly 5.5% stake in the publicly traded company. Before joining Moderna, Bancel was the CEO of French diagnostics ...
  18. [18]
  19. [19]
    Moderna Therapeutics | The Org
    Board & advisors ; Elizabeth Tallett's profile picture · Elizabeth Tallett ; François Nader's profile picture. François Nader ; Noubar Afeyan's profile picture.
  20. [20]
    Pioneering mRNA technology - Moderna
    **Summary of Moderna (https://www.modernatx.com/):**
  21. [21]
    Press release issued by Moderna, Inc. dated January 13, 2025
    Jan 13, 2025 · “We remain focused on our three strategic priorities: driving sales growth, delivering up to 10 product approvals over the next three years, and ...
  22. [22]
    Moderna sharpens focus on $1.5bn cost cutting plan after 41 ...
    Aug 1, 2025 · Moderna sharpens focus on $1.5bn cost cutting plan after 41% revenue slump. Analysts call the revenue drop driven by dipping Covid vaccine sales ...
  23. [23]
    Moderna at Morgan Stanley Conference: Strategic Path to Growth
    Sep 8, 2025 · Moderna aims to achieve cash flow break-even by 2028 through cost management and strategic growth. · Cash costs decreased from $8.9 billion in ...
  24. [24]
    HSCI-supported research leads to new class of therapeutics
    Dec 13, 2018 · Rossi's approach gets around this antiviral response by modifying the mRNA to appear like the patient's own. Once the target protein is ...
  25. [25]
    The tangled history of mRNA vaccines - Nature
    Sep 14, 2021 · In late 1987, Robert Malone performed a landmark experiment. He mixed strands of messenger RNA with droplets of fat, to create a kind of molecular stew.
  26. [26]
    Modified mRNA vaccines - Lasker Foundation
    Sep 2, 2025 · This high-profile paper woke up the scientific community to the prospects and utility of modified mRNA, and Rossi's findings inspired him to ...
  27. [27]
    This mysterious $2 billion biotech is revealing the secrets behind its ...
    Moderna aims to design messenger RNA (mRNA) that directs cells to make a protein that works as a drug or vaccine. To succeed, the mRNA has to enter the cell, ...
  28. [28]
    Building The Digital Biotech Company - Moderna
    Jun 22, 2017 · 1. Software-Like Nature of mRNA Technology · 2. Modality-Centric Platform to Ensure Repeatability · 3. Business Strategy: Massively Parallel R&D, ...
  29. [29]
    The Long History of mRNA Vaccines | Johns Hopkins
    Oct 6, 2021 · Messenger RNA, or mRNA, was discovered in the early 1960s; research into how mRNA could be delivered into cells was developed in the 1970s.
  30. [30]
    Interview with Dr. Derrick Rossi of Moderna Therapeutics
    Oct 1, 2014 · The work-around that we came up with was to modify the nucleosides. ... Derrick: Yes, that's what makes the modified-mRNA platform so ...
  31. [31]
    Challenges of Storage and Stability of mRNA-Based COVID-19 ...
    Sep 17, 2021 · The mRNA COVID-19 vaccine developed by Moderna used a proprietary ionizable lipid (SM-102) to form a stable complex (lipoplex) with the ...
  32. [32]
    [PDF] Moderna Science & Technology Day 2022
    May 17, 2022 · Moderna's proprietary vaccine LNP improves local expression and tolerability without impacting immunogenicity. Vaccine-optimized. LNP. Generic.
  33. [33]
    Our Commitment to Innovative Approaches to Delivery - Moderna
    Apr 12, 2024 · We have also invested in the development of LNPs for systemic, intramuscular, intratumoral, and pulmonary delivery of mRNA.
  34. [34]
    The Role of N1-Methylpseudouridine in COVID-19 Vaccines
    Apr 6, 2021 · A key aspect of COVID-19 mRNA vaccines is the use of the modified nucleobase N1-methylpseudouridine (m1Ψ) to increase their effectiveness.Incorporation of N1... · N1-Methylpseudouridine... · N1-Methylpseudouridine Can...
  35. [35]
    To modify or not to modify—That is still the question for some mRNA ...
    The use of m1Ψ is also considered essential to the success of the mRNA-1273 (Moderna) and BNT162b2 (BioNTech) vaccines, which were both over 90% effective ...
  36. [36]
    Engineering of the current nucleoside-modified mRNA-LNP ...
    Jul 23, 2021 · Specifically, Moderna has identified proprietary sequences for the 5'-UTR region to increase the likelihood that a ribosome bound to the 5'-end ...
  37. [37]
    Moderna's mRNA Platform
    Our mRNA platform, with its speed, scale, and flexibility, is uniquely suited to tackle current and emerging pathogens that threaten global health.Missing: LNP | Show results with:LNP
  38. [38]
    The clinical progress of mRNA vaccines and immunotherapies
    May 9, 2022 · Here we review the clinical progress of mRNA therapy as well as provide an overview and future outlook of the transformative technology behind these mRNA-based ...
  39. [39]
    mRNA vaccines for infectious diseases: principles, delivery and ...
    Aug 25, 2021 · In two separate phase I trials in 2016, Moderna evaluated two influenza candidates. These vaccines were dosed via two intramuscular injections ...
  40. [40]
    Fact Check: Moderna began clinical trials of COVID vaccine in 2020 ...
    Feb 7, 2025 · False. Moderna began human testing of its COVID-19 vaccine more than two months after the genetic sequence for the SARS-CoV-2 virus was released ...
  41. [41]
    SARS-CoV-2 mRNA Vaccine Development Enabled by Prototype ...
    Immunogenicity in mice was confirmed 15 days later. Moderna shipped clinical drug product 41 days after GMP production began, leading to the Phase 1 clinical ...
  42. [42]
    Decades in the Making: mRNA COVID-19 Vaccines | NIAID
    Apr 4, 2024 · Knowledge of mRNA and Viruses Grows · 1961 to 1990. mRNA chromosomes · 1987. NIH launches the HIV/AIDS Clinical Trials Networks. · Early 1990s.
  43. [43]
    Safety and Immunogenicity Study of 2019-nCoV Vaccine (mRNA ...
    This clinical trial is designed to assess the safety, reactogenicity and immunogenicity of mRNA-1273 manufactured by ModernaTX, Inc.
  44. [44]
    An mRNA Vaccine against SARS-CoV-2 — Preliminary Report
    Jul 14, 2020 · The mRNA-1273 vaccine induced anti–SARS-CoV-2 immune responses in all participants, and no trial-limiting safety concerns were identified.
  45. [45]
    [PDF] OPERATION WARP SPEED: Accelerated COVID-19 Vaccine ...
    Feb 5, 2021 · Moderna – Moderna's mRNA-1273 vaccine also consists of mRNA encoding ... oversight, policy, and funding decisions. GAO's commitment to ...
  46. [46]
    U.S. Raises to $2.5B toward Its “Warp Speed” Bet on Moderna's ...
    Aug 12, 2020 · The U.S. government has awarded Moderna an additional up to $1.525 billion toward manufacturing and delivering 100 million doses of the ...<|separator|>
  47. [47]
    Study Details | NCT04470427 - Clinical Trials
    This study evaluates the mRNA-1273 vaccine's efficacy, safety, and immunogenicity to prevent COVID-19, with three parts: blinded, open-label, and booster dose.
  48. [48]
    Fast Development of High-Quality Vaccines in a Pandemic - PMC
    In general, the discovery and development of a vaccine takes 8 to 20 years, with anywhere from 2 to 10 years in the clinical trial phase. Surprisingly, as of ...<|separator|>
  49. [49]
    [PDF] Moderna COVID-19 Vaccine EUA Fact Sheet for Recipients ... - FDA
    The Moderna COVID-19 vaccine options include SPIKEVAX (12+) and Moderna (12+) and Bivalent (18+) for preventing COVID-19. SPIKEVAX and Moderna can be used ...
  50. [50]
    Efficacy of the mRNA-1273 SARS-CoV-2 Vaccine at Completion of ...
    Sep 22, 2021 · The mRNA-1273 vaccine continued to be efficacious in preventing Covid-19 illness and severe disease at more than 5 months, with an acceptable safety profile.
  51. [51]
    Moderna COVID-19 Resources - Press Releases
    10/20 – Moderna Announces FDA Authorization of a Booster Dose of Moderna's COVID-19 Vaccine in the U.S. · 10/14 – Moderna Announces FDA Advisory Committee ...
  52. [52]
    [PDF] January 30, 2022 Summary Basis for Regulatory Action - SPIKEVAX
    Jan 30, 2022 · Beginning December 28, 2020, per amended protocol following the issuance of the EUA for the. Moderna COVID-19 Vaccine, all participants were ...
  53. [53]
    Moderna COVID-19 Vaccine - FDA
    On August 27, 2025, the Food and Drug Administration revoked the emergency use authorization (EUA) of Moderna COVID-19 Vaccine. Spikevax (COVID-19 Vaccine, ...
  54. [54]
    [PDF] Moderna COVID-19 Vaccine (2024-2025 Formula) Letter of ... - FDA
    Aug 22, 2024 · On August 22, 2024, FDA approved SPIKEVAX (COVID-19 Vaccine, mRNA) (2024-2025. Formula)23 for active immunization to prevent COVID-19 caused by ...
  55. [55]
    FDA Grants Full Approval to mRNA-1273 COVID-19 Vaccine in ...
    Jul 10, 2025 · mRNA-1273, marketed as Spikevax by Moderna, is now fully approved for individuals aged 6 months through 64 years with high-risk features for severe COVID-19.
  56. [56]
    Introducing mNEXSPIKE: Moderna's New COVID-19 Vaccine
    Jun 3, 2025 · mNEXSPIKE, our new COVID-19 vaccine, is now approved in the US, marking Moderna's third approved product and fifth vaccine program to achieve positive Phase 3 ...
  57. [57]
    Moderna's 2025-2026 COVID-19 Vaccines Get FDA Approval
    Sep 8, 2025 · The Food and Drug Administration (FDA) has approved the 2025-2026 formulations of Moderna's COVID-19 vaccines Spikevax® and mNexspike®.Missing: expansions 2021-2025
  58. [58]
    FDA Roundup: May 31, 2024
    May 31, 2024 · Today, the FDA approved mRESVIA (Respiratory Syncytial Virus Vaccine) ... mRESVIA is an mRNA-based vaccine that is manufactured by Moderna ...
  59. [59]
    MRESVIA - FDA
    Jun 13, 2025 · Manufacturer: ModernaTX, Inc. Indication: MRESVIA is a vaccine indicated for active immunization for the prevention of lower respiratory tract ...
  60. [60]
    MNEXSPIKE | FDA
    Sep 25, 2025 · MNEXSPIKE is a vaccine indicated for active immunization to prevent coronavirus disease 2019 (COVID-19) caused by severe acute respiratory ...
  61. [61]
    SPIKEVAX | FDA
    Sep 25, 2025 · August 27, 2025 Approval Letter - SPIKEVAX · August 26, 2025 Center Director ... OMB Approval #0910-0360 · Expiration Date 07/08/2026.
  62. [62]
    Spikevax (previously COVID-19 Vaccine Moderna)
    This was switched to a standard marketing authorisation on 3 October 2022. The name of the vaccine was changed to Spikevax on 22 June 2021.
  63. [63]
    mRESVIA FDA Approval History - Drugs.com
    Jun 13, 2025 · Approval Moderna Receives U.S. FDA Approval for RSV Vaccine, mRESVIA, in Adults Aged 18–59 at Increased Risk for RSV Disease. May 31, 2024 ...
  64. [64]
    mResvia | European Medicines Agency (EMA)
    Around 4 months after vaccination, people who received mResvia had an 84% reduction in their risk of getting lower respiratory tract disease caused by RSV, ...
  65. [65]
    Moderna receives expanded approval for RSV vaccine - CIDRAP
    Jun 13, 2025 · In a news release, Moderna said the FDA has approved mResvia (mRNA-1345) for the prevention of RSV in people ages 18 to 59 years old who are at ...
  66. [66]
    US FDA approves expanded use of Moderna's RSV vaccine for at ...
    Jun 13, 2025 · The shot, the company's second product, is already approved for the prevention of RSV-associated lower respiratory tract disease in adults aged ...
  67. [67]
  68. [68]
    Find a Trial / Results - Moderna Clinical Trials
    Trial Title. A Study to Assess the Long-term Safety and Clinical Activity of mRNA-3927 in Participants Previously Enrolled in the mRNA-3927-P101 Study.
  69. [69]
  70. [70]
  71. [71]
    A Study of mRNA-1608, a Herpes Simplex Virus - Trial Details
    A Phase 1/2, Randomized, Observer-Blind, Controlled, Dose-Ranging Study of mRNA-1608, an HSV-2 Therapeutic Candidate Vaccine, in Healthy Adults 18 to 55 Years ...
  72. [72]
    Moderna Announces Positive Phase 3 Results for Seasonal ...
    Jun 30, 2025 · mRNA-1010 demonstrated superior relative vaccine efficacy that was 26.6% (95% CI; 16.7%, 35.4%) higher than a licensed standard-dose seasonal influenza vaccine.
  73. [73]
    Moderna flu vaccine beats approved competitor in fresh phase 3 win
    Jun 30, 2025 · Moderna said its own vaccine, dubbed mRNA-1010, had achieved a relative vaccine efficacy (rVE) of 26.6% over a “licensed standard-dose seasonal influenza ...
  74. [74]
  75. [75]
    Phase IV Trial Shows Strong Immune Response from Moderna's ...
    Sep 17, 2025 · Preliminary Phase IV trial results show that Moderna's 2025–2026 Spikevax formula boosted neutralizing antibodies more than eight-fold in ...
  76. [76]
  77. [77]
    Moderna Crashes 17% After Slashing 2024-25 Guidance
    Jan 13, 2025 · Moderna (MRNA) stock crashed Monday after the Covid vaccine maker slashed its outlook for 2024 and 2025 sales and announced an expanded cost-cutting initiative.<|separator|>
  78. [78]
    Moderna Reports Fourth Quarter and Fiscal Year 2024 Financial ...
    Feb 14, 2025 · Revenue: The Company reiterates 2025 expected revenue of $1.5 to $2.5 billion. Moderna expects revenue of approximately $0.2 billion in the ...Missing: breakdown | Show results with:breakdown
  79. [79]
    Moderna Revenue 2017-2025 | MRNA - Macrotrends
    Moderna annual revenue for 2024 was $3.236B, a 52.75% decline from 2023. · Moderna annual revenue for 2023 was $6.848B, a 64.45% decline from 2022.Missing: sources breakdown
  80. [80]
    Press release issued by Moderna, Inc. dated - SEC.gov
    Aug 1, 2025 · Revenue: The Company updated its 2025 projected revenue range to $1.5 to $2.2 billion, reflecting a $300 million reduction at the high end of ...Missing: breakdown | Show results with:breakdown
  81. [81]
    How Moderna Makes Money - Investopedia
    Jun 12, 2024 · Moderna expects sales to climb in 2024 and 2025. Moderna's Industry ... Moderna Revenue Breakdown: Product sales 97%; Grant revenue 1.3%; ...
  82. [82]
    Document - SEC.gov
    Moderna is entering 2025 with two approved products, Spikevax® and mRESVIA ... Spikevax sales for the full year 2024 were $3.1 billion. RSV: The Company ...Missing: breakdown | Show results with:breakdown
  83. [83]
  84. [84]
    Moderna Gross Margin 2017-2025 | MRNA - Macrotrends
    Moderna gross margin for the quarter ending June 30, 2025 was 0%. · Moderna average gross margin for 2017-12-31 was 0%, a 0% decline from 2018-03-31. · Moderna ...Missing: breakdown | Show results with:breakdown
  85. [85]
    Financials - Moderna, Inc. (MRNA) Income Statement - Yahoo Finance
    Moderna Inc's financial performance reflects a challenging landscape, with significant revenue growth declines of -39.05% in Q2 2025, though EBITDA growth ...
  86. [86]
    Moderna Inc. (MRNA) Market Analysis: Strategic Shifts & Financial ...
    Jul 2, 2025 · The gross profit margin improved to 54.76% from 31.47% in 2023, suggesting better cost efficiencies or a shift in product mix favoring higher- ...Missing: structure | Show results with:structure
  87. [87]
    MRNA.O - | Stock Price & Latest News - Reuters
    Gross Margin (5Y), 76.34. Gross Margin (Annual), 70.06. Gross Margin (TTM), 68.32. Net Profit Margin (5Y), 23.74. Net Profit Margin % (Annual), -110.04. Net ...
  88. [88]
    Moderna Research and Development Expenses 2017-2025 | MRNA
    Moderna annual research and development expenses for 2024 were $4.543B, a 6.23% decline from 2023. Moderna annual research and development expenses for 2023 ...Missing: SG&A | Show results with:SG&A
  89. [89]
    Moderna SG&A Expenses 2017-2025 | MRNA - Macrotrends
    Moderna annual sg&a expenses for 2024 were $1.174B, a 24.21% decline from 2023. Moderna annual sg&a expenses for 2023 were $1.549B, a 36.84% increase from 2022.Missing: R&D | Show results with:R&D
  90. [90]
    Moderna Operating Expenses 2017-2025 | MRNA - Macrotrends
    Moderna annual operating expenses for 2024 were $7.181B, a 35.23% decline from 2023. · Moderna annual operating expenses for 2023 were $11.087B, a 12.64% ...Missing: breakdown | Show results with:breakdown
  91. [91]
    Moderna to Reduce Workforce by 10% Amid Cost Restructuring
    Aug 1, 2025 · 2025 revenue guidance reaffirmed: Despite a Q1 loss of $1 billion, Moderna maintains its full-year revenue outlook of $1.5 to $2.5 billion.Missing: breakdown | Show results with:breakdown
  92. [92]
    Moderna - 7 Year Stock Price History | MRNA - Macrotrends
    End of interactive chart. Moderna (MRNA) Annual % Change. 2025, -35.69%. 2024, -58.19%. 2023, -44.63%. 2022, -29.28%. 2021, 143.11%. 2020, 434.1%. 2019, 28.09% ...
  93. [93]
    Moderna, Inc. (MRNA) Stock Historical Prices & Data - Yahoo Finance
    Discover historical prices for MRNA stock on Yahoo Finance. View daily, weekly or monthly format back to when Moderna, Inc. stock was issued.
  94. [94]
    Moderna, Inc. (MRNA) Stock Price, News, Quote & History
    Find the latest Moderna, Inc. (MRNA) stock quote, history, news and other vital information to help you with your stock trading and investing.Community · Historical Data · Profile · MRNA Interactive Stock Chart
  95. [95]
    Moderna Market Cap 2017-2025 | MRNA - Macrotrends
    Stock Price History Stock Splits Market Cap. Moderna market cap as of October 25, 2025 is $10.25B. Moderna market cap history and chart from 2017 to 2025.
  96. [96]
    Moderna (MRNA) Revenue 2016-2025 - Stock Analysis
    Moderna had revenue of $142.00M in the quarter ending June 30, 2025, a decrease of -41.08%. This brings the company's revenue in the last twelve months to $3.08 ...
  97. [97]
    Investor Relations - Moderna
    **Summary of Moderna’s Investor Relations Page (as of 2025):**
  98. [98]
    Quarterly Results - Moderna Investor Relations
    Quarterly Results · Q2 2025 REPORT · Download · Related Information · Financial Summary Table · Quick Links · IR Contact · Investor Email Alerts · Investors.
  99. [99]
    [PDF] moderna-r-d-day-2024-final-presentation.pdf
    This presentation contains forward-looking statements within the meaning of the Private Securities Litigation Reform Act of 1995, as.
  100. [100]
    [PDF] Moderna COVID-19 Vaccine VRBPAC Briefing Document - FDA
    Dec 17, 2020 · On December 7, 2020, the Sponsor submitted an amendment to the EUA request with additional accrued safety data on all participants with a median ...Missing: timeline | Show results with:timeline
  101. [101]
    VAERS - Data - HHS.gov
    May 8, 2025 · VAERS is designed to rapidly detect unusual or unexpected patterns of adverse events, also referred to as “safety signals.” If a possible safety ...Guide to Interpreting VAERS... · Data Sets · Resources
  102. [102]
    Coronavirus Disease 2019 (COVID-19) Vaccine Safety - CDC
    Myocarditis and Pericarditis. Myocarditis and pericarditis after COVID-19 vaccination are rare.1234567911121718192021. Myocarditis is inflammation of the ...Myocarditis and Pericarditis · Women who would like to... · Long-term care residents
  103. [103]
    Safety and Adverse Events Related to COVID-19 mRNA Vaccines
    Cardiac complications were the most commonly reported severe adverse events. It appeared that systemic adverse reactions are more common after the second dose ...
  104. [104]
    Long-term safety and effectiveness of mRNA-1273 vaccine in adults
    Aug 29, 2024 · Primary vaccination and boosting with mRNA-1273 demonstrated acceptable safety, effectiveness and immunogenicity against COVID-19, including emergent variants.
  105. [105]
  106. [106]
    COVID-19 vaccines and adverse events of special interest
    Apr 2, 2024 · This study aimed to evaluate the risk of adverse events of special interest (AESI) following COVID-19 vaccination from 10 sites across eight countries.
  107. [107]
    Adverse Events of COVID-19 Vaccines in the United States
    Jul 15, 2024 · Objective: This study aimed to analyze the temporal and spatial characteristics of AEFIs associated with COVID-19 vaccines in the United States ...
  108. [108]
    Interim Estimates of Vaccine Effectiveness of BNT162b2 and mRNA
    Apr 2, 2021 · mRNA vaccine effectiveness of full immunization (≥14 days after second dose) was 90% against SARS-CoV-2 infections regardless of symptom status.
  109. [109]
    Real-world effectiveness of the mRNA-1273 vaccine against COVID ...
    Phase 3 trials found mRNA-1273 was highly effective in preventing COVID-19. We conducted a prospective cohort study at Kaiser Permanente Southern California ...
  110. [110]
    Vaccine effectiveness against Delta, Omicron BA.1, and BA.2 in a ...
    The primary series provided greater protection against infection with Delta (45%, 95% CI 40–50%) than against infection with Omicron (21%, 95% CI 7–34% for BA.1 ...
  111. [111]
    Evaluation of Waning of SARS-CoV-2 Vaccine–Induced Immunity
    May 3, 2023 · The estimated vaccine effectiveness against both laboratory-confirmed Omicron infection and symptomatic disease was lower than 20% at 6 months.
  112. [112]
    COVID-19 Vaccine Effectiveness Against Omicron or Delta Infection ...
    Sep 22, 2022 · After a third dose, estimated vaccine effectiveness against Omicron was 61% for symptomatic infection and 95% for severe outcomes. Meaning The ...
  113. [113]
    Covid-19 Vaccine Effectiveness against the Omicron (B.1.1.529 ...
    Mar 2, 2022 · Modest reductions in vaccine effectiveness against infection and mild disease have been observed with the beta (B.1.351) and delta variants, ...
  114. [114]
    Outcomes among patients with breakthrough SARS-CoV-2 infection ...
    We found the rate of breakthrough infection among fully vaccinated persons to be 0.66 per 1000 person-days after full vaccination [5]. However, certain ...Missing: empirical | Show results with:empirical
  115. [115]
    The Risk of Hospitalization and Mortality After Breakthrough SARS ...
    Nov 8, 2022 · Further, we found that risk of hospitalization was 40% less and risk of death was 75% less in SARS-CoV-2 breakthrough infections among ...Missing: empirical | Show results with:empirical
  116. [116]
    Waning effectiveness of mRNA COVID-19 vaccines against inpatient ...
    Mar 7, 2024 · We showed that while two doses of mRNA COVID-19 vaccine were initially highly effective, vaccine effectiveness waned as time elapsed since the second dose.
  117. [117]
    Waning effectiveness of SARS-CoV-2 mRNA vaccines in older adults
    However, accumulating evidence suggests that vaccine effectiveness (VE) against SARS-CoV-2 infection declines over time in some groups, including older adults ...
  118. [118]
    Waning of vaccine effectiveness against moderate and severe covid ...
    Oct 3, 2022 · Effectiveness of mRNA vaccines against moderate and severe covid-19 waned with time after vaccination. The findings support recommendations for a booster dose.
  119. [119]
    mRNA Patent Wars Update: The Plot Thickens with Key Rulings ...
    Mar 23, 2025 · In August 2022, Moderna sued Pfizer and BioNTech for infringing three of Moderna's patents directed to mRNA and lipid technology. While Pfizer ...Missing: details | Show results with:details
  120. [120]
    Pfizer wins pause on Moderna's COVID-19 patent lawsuit | Reuters
    Apr 12, 2024 · Moderna's patent lawsuit claiming Pfizer and BioNTech copied its COVID-19 vaccine technology will be put on hold while the U.S. Patent ...<|separator|>
  121. [121]
    Pfizer convinces US Patent Office to cancel two Moderna COVID-19 ...
    Mar 5, 2025 · A German court separately ruled earlier Wednesday that Pfizer and BioNTech infringed a European Moderna patent and owed Moderna an amount of ...
  122. [122]
    Pfizer, BioNTech lose UK appeal to overturn Moderna's COVID ...
    Aug 1, 2025 · The High Court also ruled that the other Moderna patent under challenge in the case was invalid. Moderna was refused permission to appeal ...
  123. [123]
    As COVID vaccine patent dispute drags on, Moderna forks over ...
    Feb 24, 2023 · As COVID vaccine patent dispute drags on, Moderna forks over $400M to NIH. Moderna called the sum a “catch-up payment” for borrowing a ...
  124. [124]
    Genevant Sciences and Arbutus Biopharma Initiate International ...
    Mar 3, 2025 · Genevant and Arbutus are seeking monetary relief and injunctions against Spikevax and, where applicable, additional Moderna products that Moderna has ...
  125. [125]
    Arbutus and Brinkhof celebrate interim success in mRNA case ...
    May 28, 2025 · Arbutus and Genevant jointly sued 15 Moderna companies in March 2015 at the local division The Hague for infringement of EP 2 279 254 and EP 4 ...
  126. [126]
    Moderna settles Alnylam patent lawsuits over COVID vaccine ...
    Sep 18, 2025 · Moderna has settled related lawsuits that accused it of misusing Alnylam Pharmaceuticals' patented technology in its COVID-19 vaccines, ...
  127. [127]
    Moderna fends off Alnylam US patent lawsuit over COVID shots, for ...
    Oct 2, 2024 · Alnylam Pharmaceuticals ... ALNY.O ... admitted defeat, for now, in a lawsuit in Delaware federal court that accused Moderna's ... MRNA.O ...
  128. [128]
    GSK sues Moderna for US patent infringement over COVID, RSV ...
    Oct 15, 2024 · British drugmaker GlaxoSmithKline sued Moderna in U.S. federal court in Delaware on Tuesday, accusing it of violating GSK patent rights in ...
  129. [129]
    FDA issues EUA for Moderna COVID vaccine - RAPS
    Dec 18, 2020 · The US Food and Drug Administration (FDA) on Friday authorized Moderna's mRNA-based COVID-19 vaccine for emergency use in individuals 18 and ...
  130. [130]
    Emergency Use Authorizations (EUAs) Versus FDA Approval - NIH
    EUA policies typically require data supporting—not proving—safety and effectiveness, with lower standards and faster reviews than FDA approval. Although ...
  131. [131]
    Concerns over “cosy relationship” between the FDA and Moderna
    An investigation published by The BMJ today raises concerns about a revolving door culture between the US Food and Drug Administration (FDA) and Moderna.Missing: empirical | Show results with:empirical
  132. [132]
    FDA vaccine advisers 'disappointed' and 'angry' that early data ...
    Jan 11, 2023 · Some vaccine advisers to the federal government say they're “disappointed” and “angry” that government scientists and the pharmaceutical company Moderna didn't ...
  133. [133]
    Public Returns on Public Investment: Moderna's Violation of the ...
    specifically, the value of the NIH-Moderna vaccine ...Missing: controversies | Show results with:controversies
  134. [134]
    Moderna and U.S. at Odds Over Vaccine Patent Rights
    Nov 11, 2021 · Moderna and the National Institutes of Health are in a bitter dispute over who deserves credit for inventing the central component of the company's powerful ...Missing: ethics | Show results with:ethics
  135. [135]
    COVID-19 Vaccine Patent War: Moderna's Claims Invalidated by PTAB
    Mar 7, 2025 · Previously, on August 26, 2022, Moderna sued Pfizer and BioNTech for infringing the '600 patent, the '127 patent and U.S. Patent No. 10,898,574 ...
  136. [136]
    COVID‐19 vaccine research and development: ethical issues - PMC
    Ethical issues include balancing speed with subject protection, potential exploitation of vulnerable populations, and the need for post-trial access to the  ...
  137. [137]
    The Ethics of Continuing Placebo in SARS-CoV-2 Vaccine Trials
    Both companies now claim they have an “ethical obligation” to offer vaccine as soon as possible to all participants who received placebo.Missing: controversies | Show results with:controversies
  138. [138]
    HHS Cancels Moderna Grant for Late-Stage Development of mRNA ...
    Jun 4, 2025 · HHS has pulled a $590M grant from Moderna's H5N1 mRNA flu vaccine, citing scientific and ethical concerns. Despite this, Moderna reported ...
  139. [139]
    Moderna faces audit over 'unacceptable' lack of transparency
    May 7, 2025 · Moderna is being audited by the PMCPA after yet again being found to have brought discredit on the pharma industry.Missing: data | Show results with:data
  140. [140]
    Moderna rebuked over second successive Spikevax marketing breach
    Sep 30, 2024 · In a recent PMCPA ruling, Moderna was found to have breached three clauses of the UK self-regulatory body's code.Missing: criticism | Show results with:criticism
  141. [141]
    COVID-19 mRNA vaccines: Platforms and current developments - NIH
    mRNA-1273 went through a clinical development process similar to that of BNT162b2, and a favorable safety profile and 94.1% vaccine efficacy (protection in ...
  142. [142]
    A comprehensive review of SARS-CoV-2 vaccines: Pfizer, Moderna ...
    On February 7, the first batch of the Moderna vaccine was developed and ready to use for analytical testing. The NIH was an early proponent of the mRNA-1273 ...
  143. [143]
    Clinical severity of, and effectiveness of mRNA vaccines ... - The BMJ
    Mar 9, 2022 · mRNA vaccines were found to be highly effective in preventing covid-19 associated hospital admissions related to the alpha, delta, and omicron variants.<|separator|>
  144. [144]
    [PDF] May 31, 2024 Summary Basis for Regulatory Action - MRESVIA - FDA
    May 31, 2024 · (Moderna) submitted a Biologics License Application (BLA) (STN BL. 125796/0) for licensure of their respiratory syncytial virus (RSV) vaccine.
  145. [145]
    mRNA medicines we are currently developing
    ### Ongoing Clinical Programs (as of October 23, 2025)
  146. [146]
    Medical areas of focus & Modalities - Moderna
    We are developing several potential mRNA therapeutics that we believe have the potential to help the millions of patients battling autoimmune diseases. Learn ...
  147. [147]
    How Pharma Money Colors Operation Warp Speed's Quest to ...
    Nov 30, 2020 · In total, Moderna received nearly $1 billion in federal funds for development and a $1.5 billion contract with HHS for 100 million doses.
  148. [148]
    After nearly $1B in research funding, Moderna takes $1.5B ...
    Aug 12, 2020 · At $1 billion for 100 million doses, Johnson & Johnson's Operation Warp Speed deal—plus $456 million in R&D funding—comes out to about $14.56 ...<|separator|>
  149. [149]
    [PDF] Explaining Operation Warp Speed
    How is this being funded? Congress has directed almost $10 billion to this effort through supplemental funding, including the CARES Act. Congress has also ...
  150. [150]
    US Taxpayers Heavily Funded the Discovery of COVID‐19 Vaccines
    Operation Warp Speed (OWS) invested billions of dollars in conducting rigorous clinical trials and in manufacturing with the participation of Moderna, ...
  151. [151]
    Clinical Impact and Cost-Effectiveness of Updated 2023/24 COVID ...
    The updated Moderna vaccine prevented many hospitalizations and provided cost savings, with a return on investment of $1.10–$2.60 per dollar spent.
  152. [152]
    Economic evaluation for mass vaccination against COVID-19
    Taken together, the Moderna COVID-19 vaccine yielded an average 0.8284 quality-adjusted life days (QALDs) gained per person with less cost incurred, which ...
  153. [153]
    Moderna's Global Public Health Strategy: Parsing the Hype and ...
    Mar 8, 2022 · Moderna has announced today a new Global Public Health Strategy granting only limited access to its covid vaccine patents and mRNA technology platform.
  154. [154]
    Operation Warp Speed: Accelerated COVID-19 Vaccine ...
    Feb 11, 2021 · Operation Warp Speed was a federal effort that supported multiple COVID-19 vaccine candidates to speed up development.
  155. [155]
    Comparing Moderna's mRNA-1083 and Pfizer's dual-target ... - Nature
    May 24, 2025 · Notably, the Moderna's mRNA-1083 vaccine candidate demonstrated positive results in a phase I/II clinical trial shortly after Pfizer and ...
  156. [156]
    Public health experts dismayed by RFK Jr.'s defunding of mRNA ...
    Aug 6, 2025 · The Trump administration canceled about $500 million for research into mRNA vaccines. The move slows progress in using the technology to ...Missing: influences | Show results with:influences
  157. [157]
    mRNA, once lauded as a scientific marvel, is now a government target
    Jun 2, 2025 · The sharp turnaround on mRNA vaccines shows an unprecedented rise of anti-vaccine activists to power and an attack on accepted science.Missing: influences | Show results with:influences
  158. [158]
    Moderna Recognizes the Success of Operation Warp Speed and ...
    Sep 3, 2025 · Since 2021, the real-world effectiveness and safety of the vaccines have been confirmed by governments and health systems around the world.Missing: 1273 funding
  159. [159]
    6 in 10 Americans do not plan to get updated COVID vaccine for ...
    Nov 19, 2024 · Six-in-ten Americans say they will probably not get an updated 2024-25 COVID-19 vaccine, according to an October Pew Research Center survey.
  160. [160]
    KFF Tracking Poll on Health Information and Trust: COVID-19 ...
    Aug 1, 2025 · The Fall 2025 COVID-19 Vaccines​​ Overall, most of the public (59%) say they will either “definitely not” or “probably not” get the COVID-19 ...
  161. [161]
    Bancel, Sanders spar over what Moderna owes the government
    Mar 22, 2023 · Moderna is planning to price its vaccines at about $130 per dose, after selling the products for significantly less to the federal government.
  162. [162]
    Moderna COVID-19 Vaccine | ACIP - CDC
    About 22% of vaccine recipients and 4% of placebo recipients reported any grade ≥3 local or systemic reactions following either dose 1 or dose 2. The Phase III ...
  163. [163]
    A descriptive review on the real-world impact of Moderna, Inc ...
    Expert opinion: Post-marketing safety and real-world data for Moderna, Inc. COVID-19 mRNA vaccines strongly support a positive benefit - risk profile favoring ...
  164. [164]
    What we know about the safety, efficacy of mRNA vaccines amid ...
    May 23, 2025 · Infectious disease experts say mRNA vaccines have been studied for decades, they are safe and effective, and were instrumental in saving ...
  165. [165]
    Safety outcomes associated with the moderna COVID-19 vaccine ...
    Expert Opinion. The transient nature of commonly observed AEs and the rare occurrence of severe events within mRNA-1273 recipients show no significant safety ...
  166. [166]
    Lessons Learned from Unfettered U.S. Government Support of the ...
    The NIH-Moderna mRNA COVID-19 vaccine's steep price increase raises concerns that this will be the new anchor for continued price hikes.