Blog

  • Court of Arbitration’s latest award on Indus Waters Treaty vindicates Pakistan’s stance — FO

    Court of Arbitration’s latest award on Indus Waters Treaty vindicates Pakistan’s stance — FO

    ‘Landmark deal’: Pakistan’s stock market gains on optimism over US trade negotiations


    ISLAMABAD: Pakistan’s stock market maintained its bullish momentum on Monday, buoyed by reports of potential US investment in the energy sector and comments from the state finance minister that Islamabad and Washington would fine tune the details of a trade pact in the months ahead.


    The KSE-100 Index climbed past the 147,000 points mark during intraday trading and closed at 146,929.84, up 1,547.05 points, or 1.06 percent, from Friday’s close of 145,382.79.


    Positive investor sentiment has been underpinned by US President Donald Trump’s 19 percent tariffs on Pakistani imports announced last month, which officials say will pave the way for renewed investment by American firms and deepen economic ties between the two countries.


    Topline Securities, a Karachi-based brokerage, said market giants like Mari Petroleum Company (MARI), Bank AL Habib Limited (BAHL), Oil and Gas Development Company (OGDC), Meezan Bank Limited (MEBL) and Muslim Commercial Bank (MCB) dominated Monday’s rally, collectively adding 959 points to the index.


    “Sentiment surged after reports of US firms gearing up to invest in Pakistan’s energy sector, further reinforced by better-than-expected corporate results that added to the market’s upbeat tone,” the report said.


    The total traded volume reached 607 million shares with a trading value of Rs43.95 billion. Lotte Chemical Pakistan Limited (LOTCHEM) led the volumes chart, with 73 million shares changing hands.


    Market analysts say the positive momentum reflects growing investor confidence in Pakistan’s economic prospects, helped by strengthened US ties that are expected to support further gains in the near term.


    Pakistan’s State Minister for Finance, Bilal Azhar Kayani, described the US trade pact as a “landmark” deal, saying the 19 percent tariff was the lowest in the South Asian region.


    “And the agreement with more details will be negotiated and discussed in the months ahead,” he said during an interview with Bloomberg.


    “Which would include various aspects, rules of origin or market access or tariffs per specific lines reciprocally.”


    Kayani noted that the US was Pakistan’s largest export destination, accounting for $6 billion of the country’s $32 billion in exports last fiscal year.


    Pakistan’s exports to the US are dominated by textiles and garments, but also include leather goods, surgical instruments, sports equipment, chemicals, carpets and seafood, according to the Ministry of Commerce.


    The new trade agreement comes amid signs of a thaw in relations between Islamabad and Washington after years of friction over security and counterterrorism. The Biden administration maintained a cautious approach toward Pakistan, but Trump has spoken warmly of his interactions with Pakistani officials, including an unprecedented two-hour meeting in June with the Pakistan army chief. More recently, US officials have emphasized trade and investment cooperation, particularly in crypto, energy, textiles, and information technology sectors.

    Continue Reading

  • Meet the Real-Life Astronauts Portrayed in the Movie

    Meet the Real-Life Astronauts Portrayed in the Movie

    Key Takeaways:

    • Astronaut Jim Lovell, who died Thursday, was part of the three-man crew on the Apollo 13 NASA mission in 1970.
    • Decades later, Lovell wrote a book about the near-disaster that inspired the Oscar-winning movie Apollo 13.
    • Lovell and his crew members, Fred Haise Jr. and Jack Swigert, had decorated careers within the space agency. Today, Haise is the only living member of the famous mission.

    “Houston, we have a problem.” The iconic five-word phrase spoken by Tom Hanks, portraying astronaut Jim Lovell, in the 1995 blockbuster Apollo 13 instantly became one of the most memorable movie quotes of all time. However, the line was one instance where director Ron Howard’s Academy Award–winning movie took some creative license.

    Astronaut Jack Swigert, played on the big screen by Kevin Bacon, was actually the one who first sent the famous distress call to Mission Control from the shuttle, saying, “Okay, Houston, we’ve had a problem here.” (Lovell repeated the phrase when a capsule communicator in Houston asked what Swigert had said.) Other than that slight change of phrase, Apollo 13 was a very real depiction of the perilous outer space journey of three astronauts: Lovell, Swigert, and Fred Haise (Bill Paxton).

    Intended to be NASA’s third moon-landing mission, Apollo 13 launched from Florida’s Kennedy Space Center on April 11, 1970. Two days later—and approximately 205,000 miles from Earth—the men reported hearing a loud bang. That problem Swigert and Lovell had famously mentioned ended up being an oxygen tank explosion that severely damaged the vessel, rendering a lunar landing impossible.

    “It really was not until I looked out the window and saw the oxygen escaping from the rear end of my spacecraft that I knew that we were in serious trouble,” Lovell recalled at a Kennedy Space Center gala commemorating the mission’s 45th anniversary in April 2015.

    Across the nation, Americans were glued to their television sets awaiting news of what happened to the trio who was forced to orbit the moon while scrambling to find a way to back to their families. After spending 142 hours and 54 minutes in space, the crew returned safely to Earth on April 17. They landed in the south Pacific Ocean, about four miles from the recovery ship, USS Iwo Jima.

    The following day, President Richard Nixon awarded the Presidential Medal of Freedom to the men, as well as Apollo 13’s Mission Operations Team for their heroic efforts during what has been called “NASA’s finest hour.”

    These are the real men behind the famed space mission and hit film:

    Commander James “Jim” Lovell Jr.

    Portrayed by Tom Hanks

    Getty Images

    Astronaut Jim Lovell, left, and Tom Hanks portraying Lovell in Apollo 13

    With three missions and 572 spaceflight hours of experience to his credit, Lovell was the world’s most traveled astronaut for a time. A former test pilot, the Ohio native participated in several high-profile NASA missions, including flights on Gemini 7, Gemini 12, and Apollo 8, which was the first mission to circle the moon.

    Before joining NASA, Lovell attended the United States Naval Academy in Annapolis, Maryland, from 1948 to 1952. Upon graduation, he married his high school sweetheart Marilyn Lovell (née Gerlach), whom actor Kathleen Quinlan portrayed in Apollo 13. The couple had four children: Barbara, James III, Susan, and Jeffrey.

    Lovell’s role as Apollo 13 commander is one he’s often reflected on over the years. “The flight was a failure in its initial mission,” he said at the 2015 Kennedy Space Center gala. “However, it was a tremendous success in the ability of people to get together, like the mission control team working with what they had and working with the flight crew to turn what was almost a certain catastrophe into a successful recovery.”

    On March 1, 1973, Lovell retired from NASA and as a U.S. Navy captain. After working in various corporate jobs, including executive roles in a towing company and telecommunications business, he retired from the private sector in 1991. In collaboration with journalist Jeffrey Kluge, Lovell co-wrote the 1994 book Lost Moon: The Perilous Voyage of Apollo 13, which served as the basis for Howard’s big-screen adaptation the following year. Lovell even made a cameo in the movie as captain of the USS Iwo Jima rescue ship. He died Thursday at age 97.

    Lunar Module Pilot Fred Haise Jr.

    Portrayed by Bill Paxton

    portrait of a man in a blue uniform smiling and holding a model space shuttle

    Getty Images

    Astronaut Fred Haise Jr.

    a man in a white outfit peers outside a window partially covered by frost

    Getty Images

    Bill Paxton as astronaut Fred Haise Jr. in Apollo 13

    Born in Biloxi, Mississippi, Haise completed flight training with the U.S. Navy in 1954 and served in the U.S. Marine Corps as a fighter pilot until 1956. Beginning his NASA career in 1959 at the Lewis Research Center (now the Glenn Research Center) in Cleveland, the University of Oklahoma graduate acted as a research pilot until he was selected for astronaut training at Houston’s Johnson Space Center in 1966.

    Now 91, Haise had been a backup lunar module pilot on Apollo 8 and 11, but his earlier experience proved most useful in calmly helping the Apollo 13 crew survive the aborted lunar-landing mission. “As a military pilot and a test pilot, handling unusual situations and aircraft malfunctions was part of the business,” he explained in a 2014 Q&A with NASA. “My biggest emotion on Apollo 13 after the oxygen tank explosion was disappointment that we had lost the landing. Ron Howard, director for the movie Apollo 13, commented that it never sounded like we had a problem after listening to all the air-to-ground transmissions.”

    He was later assigned to command the Apollo 19 moon mission that NASA ultimately canceled in 1972 following a series of budget cuts. Along with fellow astronaut Gordon Fullerton, Haise piloted the space shuttle Enterprise for three of its test flights in 1977. After leaving NASA two years later, the father of four served as president of Grumman Technical Services Inc. as part of the Shuttle Processing Contract Team throughout the 1980s and ’90s until his eventual retirement.

    Command Module Pilot John “Jack” Swigert Jr.

    Portrayed by Kevin Bacon

    a man in astronaut suit smiles for a photo portrait behind a desk

    Getty Images

    Astronaut Jack Swigert

    a man in an astronaut suit stands and looks over his shoulder to the right

    Getty Images

    Kevin Bacon as astronaut Jack Swigert in Apollo 13

    Swigert was a last-minute addition to the Apollo 13 crew, replacing Ken Mattingly, who had been exposed to German measles just 48 hours before the 1970 launch. The Denver native served in the United States Air Force from 1953 to 1956 and was assigned as a fighter pilot in Japan and Korea upon his graduation from the Pilot Training Program and Gunnery School at Nellis Air Force Base in Nevada.

    Following his tour of active duty, Swigert served as a jet fighter pilot in both the Massachusetts Air National Guard from 1957 to 1960 and the Connecticut Air National Guard from 1960 to 1965. In April 1966, Swigert and Haise were among the 19 astronauts selected by NASA for training, and two years later, the former became a member of Apollo 7’s astronaut support crew. The Apollo 13 mission was the then-38-year-old mechanical and aerospace engineer’s first space flight.

    After taking a leave of absence in April 1973 to become the U.S. House of Representatives’ Executive Director of the Committee on Science and Technology, Swigert eventually resigned from both NASA and the congressional committee in August 1977 to officially enter politics. The Republican was elected to the U.S. House of Representatives from Colorado’s 6th district in 1982. Before he could be sworn in, however, Swigert died of bone cancer in December 1982 at age 51.

    Continue Reading

  • Lily James Plays Bumble CEO in Dating App Movie

    Lily James Plays Bumble CEO in Dating App Movie

    The first trailer for 20th Century Studios’ upcoming biopic movie “Swiped” has been revealed. The film will have its debut on Sept. 19 on Hulu in the U.S. and Disney+ in other territories. 

    “Swiped” introduces recent college grad Whitney Wolfe Herd, played by Lily James, as she uses grit and ingenuity to break into the male-dominated tech industry and launch an innovative, globally lauded dating app. The film will follow the rise of the online dating platform Bumble, and Wolfe’s journey to becoming the youngest female self-made billionaire. “Swiped” will touch on Wolfe’s time at Tinder as a co-founder, and the moments leading up to Bumble going public in 2021.

    The film also stars Jackson White, Myha’la, Ben Schnetzer, Pierson Fodé, Clea DuVall, Pedro Correa, Ian Colleti, Coral Peña, Dan Stevens, Larkin Woodward, Ana Yi Puig, Olivia Rose Keegan, Joley Fisher, and Gabe Kessler.

    “Swiped” is directed by Rachel Lee Goldenberg (“Unplanned,” “Minx,” “Valley Girl”) and is written by Goldberg, Bill Parker, and Kim Caramele. The film is produced by James, Jennifer Gibiot, Andrew Panay, Gala Gordon, and Sarah Shepard. 20th Century Studios and Ethea Entertainment are producing the film, with Hulu set to distribute. The film will be scored by Chanda Dancy. Doug Emmett will serve as the film’s cinematographer, with Julia Wong as editor.

    “Swiped” is set to premiere at the Toronto International Film Festival in the Gala Presentations section, and will have its world premiere on Sept. 9. 

    Watch the trailer for 20th Century Studios’ “Swiped” below.

    Continue Reading

  • MGK Addresses Sydney Sweeney Dating Rumors, Boy Band Collab Dream

    MGK Addresses Sydney Sweeney Dating Rumors, Boy Band Collab Dream

    Between being a new dad to baby daughter Saga Blade and absolutely burning the midnight oil promoting his new Lost Americana album you’d imagine MGK doesn’t have a ton of free time. Which might be a partial explanation for why the hard-charging rapper-turned-pop-punker had an exasperatedly short answer to a fan question during Sunday night’s (Aug. 10) Watch What Happens Live with Andy Cohen.

    Asked if there was any truth to the rumor that he and Euphoria star Sydney Sweeney were “more than just friends,” Kelly looked straight at the camera, shook his head and smiled, “Kyle P… shut up dude.”

    You’d think that Kelly would be content when it comes to dream collaboration wish-fulfillment, given that none other than Bob Dylan provided the voice-over for the Lost Americana trailer. But another fan question spurred MGK to reveal that he has another unexpected name on his to-do list.

    Asked if he could collab with any musician, living or dead, who he has not yet gotten in the studio with, MGK first went with late crooner Frank Sinatra. “I like Frank,” MGK said, before noting that he’s me AJ McLean from the Backstreet Boys before and considered asking them if he could hop on stage with the boy band during one of their shows at Las Vegas’ Sphere. “‘Let me hop in on the ‘[Everybody]Backstreet’s Back’ dance or something,’” he thought about asking.

    Why didn’t it happen? “Because I never sent a message or something,” Kelly admitted about his failure to get in on the BSB shows at the Sphere, which kicked off last month and include another run of gigs this weekend (Aug. 15-17).

    During a round of “Textual Behavior,” MGK also confirmed that he doesn’t have a finsta account, but is intrigued by them, while fully suggesting, Mariah-like, that he doesn’t know if his chronological age even “exists” or, if he’s being honest, have much information about his life.

    “Like if my skin rips open it heals really quick,” he said cryptically. Asked if he might have some “otherworldly” qualities, Kelly said he did ask his mom one time if she recalled going “missing” at any point or if a “tall, slender creature” ever visited her, casually mentioning that his mother did once say she thought she’d been abducted by extraterrestrials.

    Watch MGK on WWHL below.

    Continue Reading

  • ESPN, Fox to bundle upcoming streaming services in new sports-heavy deal – Reuters

    1. ESPN, Fox to bundle upcoming streaming services in new sports-heavy deal  Reuters
    2. ESPN, Fox to bundle upcoming streaming services for $39.99 a month  CNBC
    3. Strategic Synergy and Revenue Catalysts in Streaming: A New Era for Sports Media Investment  AInvest
    4. ‘Fox One’ Standalone SVOD Service Launching Aug. 21, Priced at $19.99 Monthly  Media Play News
    5. ESPN DTC and FOX One to Launch Combined Bundle Offer  The Walt Disney Company

    Continue Reading

  • How to Watch the 2025 Standard Portland Classic – LPGA

    How to Watch the 2025 Standard Portland Classic – LPGA

    1. How to Watch the 2025 Standard Portland Classic  LPGA
    2. The Standard Portland Classic  KATU
    3. 2025 The Standard Portland Classic field: LPGA Tour players, rankings  Golf News Net
    4. The Standard Portland Classic women’s golf tournament returns for 53rd year  The Business Journals
    5. The Standard Portland Classic Welcomes Top Women Golfers Aug. 14 – 17, 2025  Business Wire

    Continue Reading

  • Immunological study provides new insights into post-pandemic return of respiratory viruses

    Immunological study provides new insights into post-pandemic return of respiratory viruses

    COVID-19 prevention methods such as masking and social distancing also suppressed the circulation of common respiratory diseases, leaving young children lacking immunity to pathogens they otherwise would have been exposed to, a new multicenter clinical research study reveals. The investigators say their findings help explain the large post-pandemic rebound in these diseases and enable more accurate predictions for the future.

    The study, published Aug. 6 in The Lancet Infectious Diseases and funded by the National Institutes of Health, followed 174 children under the age of 10 from 2022-23 across four academic medical centers across the country: Weill Cornell Medicine; University of Colorado Anschutz Medical Campus and Children’s Hospital Colorado; University of North Carolina; and the University of Alabama, Birmingham. Through repeat blood sampling and respiratory sampling during illness, the investigators gauged the children’s level of immunity to many common and emerging respiratory viruses, such as RSV (respiratory syncytial virus), influenza and enterovirus D68 (EV-D68), which can cause the polio-like illness acute flaccid myelitis.

    The findings, among the first from the National Institutes of Health’s PREMISE (Pandemic Response Repository through Microbial and Immune Surveillance and Epidemiology) Program, showed that most young children lacked immunity to many normal respiratory viruses during the pandemic, suggesting they had not been exposed, as they typically would have, due to prevention measures in place. Enrolled children received routine medical care while participating in the observational study. Following the lifting of pandemic measures, the level of immunity rose across all pathogens studied, reflective of the unprecedented widespread resurgence of these viruses in children post-pandemic.

    “PREMISE is a one-of-a-kind study as we followed very young children, with their parents’ consent, over a year for longitudinal sample collection, affording us the unique opportunity to assess immunity due to primary infection, re-exposure and even vaccination, during a time when mask requirements and other nonpharmaceutical interventions were lifted,” said co-first author Dr. Perdita Permaul, section chief of pediatric allergy and immunology, associate professor of clinical pediatrics and trial principal investigator at Weill Cornell Medicine.

    The data allowed experts to recreate past circulation patterns and model predictions for future outbreaks with greater accuracy and precision. They showed that PREMISE data from 2022-23 could be used to accurately predict the subsequent wave of disease of the emerging pathogen EV-D68 that occurred in 2024.

    “Findings from our study successfully demonstrate the utility of longitudinal immunologic surveillance in children, particularly young immunologically naïve unexposed children, to help model the behavior of endemic viruses,” said Permaul, who is also an Englander Clinical Scholar at Weill Cornell Medicine and a pediatric allergist and immunologist at NewYork-Presbyterian Komansky Children’s Hospital of Children’s Hospital of New York.

    Investigators have so far evaluated nearly 1,000 children through PREMISE, based at NIH’s Vaccine Research Center, providing a treasure trove of sampling and data that can be used to learn which parts of viruses the human immune system attacks to develop immunity. This information may enable teams to better design new antibody treatments and effective vaccines to mimic this response.

    “This approach allows for the rapid development of vaccine and monoclonal antibody therapeutics for pathogens of interest in children,” Permaul said. “Future analysis of blood samples collected from almost 1,000 children enrolled in PREMISE includes pathogen-specific T and B cell responses. Longitudinal immune surveillance in young children is an important tool for informing public health planning, assessing the effectiveness of pharmacologic and non-pharmacological interventions, developing ‘on the shelf’ therapeutics and mitigating overall disease burden.” 

    This study was funded by a subcontract with Frederick National Laboratory for Cancer Research (FNLCR), currently operated by Leidos Biomedical Research, Inc. through Agreement 21X192QT1. FNLCR funding was provided by the NIH’s Vaccine Research Center. The total project funding is $7.98 million over five years. No financing for this project is supplied by nongovernmental sources.

    A version of this story first appeared on Children’s Hospital Colorado’s Newsroom.

    Alyssa Sunkin-Strube is assistant director of editorial at Weill Cornell Medicine.

    Continue Reading

  • Individuals Recovered From Severe Covid-19 Are Predispose To Develo

    Individuals Recovered From Severe Covid-19 Are Predispose To Develo

    Introduction

    After first emerging in Wuhan, China, a pandemic was swiftly turned into a novel coronavirus (SARS-CoV-2) toward the end of 2019 and caused severe impacts on health and economic systems worldwide. SARS-CoV-2 is also named COVID-19 and affects the respiratory system most, but the disease effects are not limited to the lungs. Venous thromboembolism, acute kidney and liver damage, the release of cytokines, sepsis, disseminated intravascular coagulation, complications of pregnancy, and cardiac and neurological issues are all possible outcomes of a COVID-19 infection, in addition to pulmonary involvement.1

    According to a report published in Wuhan, China, arrhythmia was found in 16.7% of hospitalized patients with COVID-19 and 44.4% of those monitored in intensive care.2 Although some studies conducted to date have revealed some clues about the potential for arrhythmia in individuals who have active disease, as of today, we are far from being able to explain why individuals who have completely recovered from the disease apply to outpatient clinics with complaints of palpitations.3,4

    Heart rate turbulence (HRT) originates from the principle of accelerating sinus rhythm, which occurs as a reflex due to the short diastole period during ventricular premature beat in patients with sinus rhythm. In a healthy heart, there is an acceleration in ventricular rate and there is a degree of this acceleration. HRT indicates this degree of acceleration. In other words, it shows the autonomic activity of the heart. For an healthy HRT both the sympathetic and parasympathetic pathways of the heart must be intact.5 Abnormal HRT, which can occur due to many diseases, indicates decreased baroreflex sensitivity and autonomic dysfunction. It is well established that individuals with blunted HRT are more likely to die suddenly and from all causes.6

    It has been revealed by the recent research on holter and pace that HRT is also impaired after atrial premature stimulation. Impaired HRT values, suggesting that the spontaneous emergence of clinical atrial fibrillation is associated with a brief increase in vagal outflow following early atrial excitement.7,8

    Conditions where a range of symptoms persist for more than three months after recovery from acute COVID-19 are called Long-COVID.9 Long-COVID-19 may manifest itself with symptoms, such as chest pain, palpitations, cognitive disorders, breath shortness, fatigue, attention deficit, hair loss, headache, myalgia, and arthralgia.9–12

    Recently, there has been an increase in the admission of patients who recovered from COVID-19 to the clinic complaining of palpitations and that these patients are referred to another departments with the preliminary diagnosis of somatization or panic disorder. Could a more sensitive assessment be made for these individuals? Could SARS-CoV-2 cause permanent damage to the autonomic nervous system (ANS) and predisposition to atrial fibrillation? This study utilized HRT to analyze the persistent impacts of SARS-CoV-2 on cardiac autonomic function in individuals recovered from COVID-19.

    Materials and Method

    Study Population

    This retrospective study analyzed the records of 10,081 participants who visited the cardiology outpatient clinics at Elazığ Fethi Sekin City Hospital between June 1, 2022, and December 31, 2024, and underwent a 24-hour ECG Holter monitoring.

    Inclusion Criteria

    A total of 328 patients with a history of positive SARS-CoV-2 test results by at least one Real-Time (RT)-PCR test on nasopharyngeal swabs (Bio-Rad CFX96 RT PCR Detection System, Bio-Rad Laboratories, Inc., Hercules, CA, USA) were included in the Recovered COVID-19 group. A total of 407 individuals with no history of positive (RT)-PCR test results for SARS-CoV-2 during the same time period and no cardiac or systemic disease (other than hypertension) detected by physical examination, laboratory tests, or anamnesis were included in the control group (Figure 1).

    Figure 1 The Subjects inclusion flowchart diagram.

    Exclusion Criteria

    Subjects with inadequate data in their files, were under 17 years old, those with atrial fibrillation, electrolyte imbalance, structural heart disease, heart valve disease, or systemic disease (apart from hypertension), individuals using anti-arrhythmic medications or agents that may lead to arrhythmia (such as terfenadine, probucol, erythromycin, amiodarone, antidepressants, clarithromycin, and antipsychotics), professional athletes, patients with a body mass index (BMI) above than 35, and pregnant women were excluded.

    Collection of Data for the Post COVID-19 Period

    SARS-CoV-2 was caught at least once and no more than three times by the subjects in the COVID-19 recovery group. Individuals in the recovered COVID-19 group had a minimum of 4 weeks and a maximum of 204 weeks between 24-hour ECG-Holter evaluation and their most recent positive RT-PCR test.

    Analysis of COVID-19 Severity with Thorax CT

    The recovered COVID-19 group was given a semi-quantitative chest CT severity score for each of the five lung lobes (two on the left and three on the right). Visual assessment was used to determine each lobe’s percentage of involvement. The chest severity score was obtained by adding the scores corresponding to the percentage of involvement of each lobe (Table 1).5

    Table 1 Semi-Quantitative Chest Computed Tomography (CT) Severity Score and Chest CT Severity Index

    Since COVID-19, which accompanies conditions such as chronic obstructive pulmonary disease and myocardial infarction, can cause severe respiratory distress and arterial blood values <90%, patients with concomitant systemic diseases were excluded from the study before their chest tomography was analyzed. Depending on their chest CT severity score, the recovered COVID-19 group was separated into three subgroups. Mild, moderate, and severe were the classifications assigned to group II (155 instances), group III (56 patients), and group IV (42 patients). Group II (modest): The CT severity score was classified as modest (<8). Group III (Moderate): The CT severity score fell within the moderate range.8–15 Group IV (Severe): According to Table 1, the CT severity score was rated as severe.16–25

    The current research was carried out in compliance with the Declaration of Helsinki’s tenets. The Ethics Committee of T.C. Fırat University granted the ethical permission (No: 2021/12-45). In compliance with the confidentiality and compliance of patient data, patient consent was waived by the ethics committee due to the retrospective nature of the study and therefore patient consent was not obtained.

    Description of Supraventricular Ectopic Beats

    To describe supraventricular ectopic beats, R-R interval tachograms and 4-lead Holter recordings were simultaneously examined. If, in addition to the presence of definitive evidence of abnormal atrial depolarization, the R-R interval was shortened by at least 20%, an ectopic beat was classified as a supraventricular premature beat. Only isolated premature supraventricular ectopic beats with a significant post-ectopic pause were evaluated.7

    24-Hour ECG-Holter Monitoring

    To evaluate HRT, 24 hour electrocardiography (ECG)-Holter data were evaluated using a 4-lead Holter device (Digitrak XT, Philips Medical Systems, Andover, USA) and Cardioscan II premier software (firmware version C.2). HRT parameters were measured by the method reported by Bauer et al.6 Turbulence onset (TO), which represents the beginning the sinus rhythm acceleration phase, and turbulence slope (TS) which describes the deceleration phase, were the two numerical IDs employed for the measurement. The start of heart rate turbulence (HRT) was defined as the difference, expressed as a percentage, between the mean of the last 2 sinus rhythm RR intervals before the supraventricular ectopic beat and the mean of the first two sinus rhythm RR intervals following the compensatory pause following the supraventricular ectopic beat. HRT onset was computed with the help of the formula below:


    RR −1 and RR-2 refer to the 2 RR intervals immediately before the supraventricular ectopic beat, and RR1 and RR2 refer to the 2 RR intervals immediately after the compensatory pause (Figure 2). The highest positive slope of a regression line assessed across any five consecutive sequences in the first fifteen consecutive sinus intervals following a supraventricular ectopic beat was defined as the turbulence slope (TS), expressed in milliseconds per beat. An HRT Slope of ≤2.5 ms/beat and an HRT Onset of ≥0% were considered abnormal.7

    Figure 2 Measurement of the interval between RR1, RR2 and RR −1, RR-2 for supraventricular ectopic beats from 24 Hour ECG-Holter.

    Statistical Evaluation

    SPSS software (SPSS Inc., Chicago, IL, USA) version 27.0 was used for statistical analysis. The Kolmogorov–Smirnov test was employed to examine the ability of continuous variables to follow a normal distribution. Since all of the continuous variables showed abnormal distribution, for the one-way ANOVA test, Tamhane’s T2 correction was employed to analyze these parameters and the variables were shown as median with 25th–75th. For categorical variables, using the chi-square test, variations between groups in baseline characteristics were evaluated and were presented as numbers and percentages. Spearman’s Rho and Pearson’s correlation analysis were used to analyze the correlation between continuous variables. Binary logistic regression analysis was conducted to detect which variables would independently predict the presence of abnormal atrial HRT onset. Results were presented as hazard ratios and 95% CI. Linear regression analysis was utilized to detect which variables would independently affect atrial HRT onset value. Results were presented as unstandardized and standardized β coefficients. P values were always two-tailed, and statistical significance was defined as values below 0.05.

    Results

    Of the 10081 cases whose 24-hour ECG Holter recordings were retrospectively examined, 735 (The controls: 407, The recovered COVID-19 group: 328) were taken in the research. All cases were separated into four groups (controls, recovered mild COVID-19, recovered moderate COVID-19, recovered severe COVID-19). Although the prevalence of abnormal atrial HRT Onset existence and atrial HRT Onset value were significantly higher in the recovered severe COVID-19 group compared to the other groups, no difference was found between the control and other Recovered COVID-19 groups (Table 2) (Figure 3). However, no difference was found among the control group and other Recovered COVID-19 groups in terms of abnormal atrial HRT Slope prevalence, atrial HRT Slope value, HT, smoking, age, and gender (Table 2).

    Table 2 Comparison of Atrial HRT Parameters Between Recovered COVID-19 Subgroups and the Control Group

    Figure 3 Comparison of Atrial HRT Onset values between the groups.

    Spearman’s rho and Pearson’s correlation analyses revealed a positive correlation between atrial HRT Onset and recovered COVID-19 subjects’ chest CT severity score and recovered COVID-19 subgroups, while no association was found between atrial HRT Onset and the Number of positive PCR tests for COVID-19 and time elapsed after COVID-19 (Table 3) (Figures 4 and 5).

    Table 3 Spearman’s Rho and Pearson’s Correlation Analyses Between Atrial HRT Onset Value and Some Other Variables

    Figure 4 Spearman’s rho correlation analyses between Atrial HRT Onset value and groups.

    Figure 5 Pearson’s correlation analyses between Atrial HRT Onset value and recovered COVID-19 subjects’ chest CT severity score.

    Regression analyses revealed that recovery from severe COVID −19, recovered COVID-19 subjects’ chest CT severity score, HT and smoking were predictors for abnormal atrial HRT onset existence and independently affected atrial HRT onset values (Table 4 and Table 5).

    Table 4 Model-1. Binary Logistic Regression for Variables (Dependent Variable: Abnormal Atrial HRT Onset Existence)

    Table 5 Model-2. Linear Regression for Variables (Dependent Variable: Atrial HRT Onset Value)

    It is clear from the table that the logistic model was developed to ascertain if the independent variables are useful in forecasting the existence of abnormalities. The model’s variables have a 19.7% explanatory power in predicting the existence of aberrant atrial HRT onset, and atrial HRT onset is significant (Model-1. p<0.001; Nagelke R2 = 0.197) (Table 4). As can be seen from the table, having a history of severe COVID-19 is a 3.851-fold risk factor for abnormal atrial HRT Onset existence compared to not having a history of severe COVID-19 (p=0.002).

    The developed linear regression model is significant when Model-2 is analyzed, and its explanatory power for the impacts of its variables on the atrial HRT Onset value is 9.2% (p<0.001; Nagelke R2 = 0.092) (Table 5). The table shows that the recovered COVID-19 subjects’ chest CT severity value variable has a statistically significant effect on the abnormal HRT Onset value. In other words, a 1-point increase in chest severity scores of individuals recovering from COVID-19 increases the atrial HRT Onset value by 0.033 units (p<0.001).

    Discussion

    This study revealed that atrial HRT values and the presence of abnormal atrial HRT onset were higher in individuals who had severe COVID-19 and recovered than in other individuals who did not have, while the atrial HRT slope value and the presence of abnormal atrial HRT slope did not differ between the groups (Table 2) (Figure 3). Although a positive connection was found among atrial HRT onset value/abnormal atrial HRT onset presence and subjects’ chest severity score and COVID-19 subgroups with correlation analyses, no association was detected between these parameters and having a positive PCR test record for SARS-CoV-2 and the time elapsed since COVID-19 (Table 3) (Figures 4 and 5). Furthermore, analyses of regression showed that recovered COVID-19 subjects’ chest CT severity score, recovering from severe COVID-19, smoking and HT were independent predictors of atrial HRT Onset value and abnormal atrial HRT onset presence (Table 4 and Table 5).

    SARS-CoV-2 binds to cardiomyocytes, type 2 pneumocytes, macrophages, and perivascular pericytes by binding to transmembrane angiotensin-converting enzyme 2 (ACE2) following proteolytic cleavage of the S-protein by serine protease. ACE2 is required for viral invasion, and ACE2 protein is found in many tissues, including the myocardium and central nervous system (CNS).13,14 In addition to direct viral invasion of myocardial and coronary endothelial cells, systemic inflammation, inappropriate T helper cell response after cytokine storm, increased calcium in myocytes induced by hypoxia causing apoptosis, hypoxia due to cardiac and respiratory failure, increased adrenergic stimulation and increased endogenous stress hormones, electrolyte imbalance, and side effects of medications taken to treat COVID-19 may be shown among the causes of cardiac clinical pictures seen in COVID-19.15,16 Stromal edema in the heart, secondary to vasculitis resulting from monocyte and lymphocyte infiltration into arterial and venous endothelial cells, is considered another cause.17 It was thought that SARS-CoV-2 had neural invasion based on genomic sequencing from cerebrospinal fluid (CSF) obtained from patients diagnosed with encephalitis and the isolation of similar viruses in brain tissue during autopsy studies.18,19 Considering that ACE2 is found in the capillary endothelium, it is thought that COVID-19 can reach the CNS by damaging the blood-brain barrier in this way.20 Some studies suggest that SARS-CoV-2, like previously reported SARS-CoV, may pass through the cribriform plate of the ethmoid bone into the systemic circulation and the virus may perform central invasion through the microcapillary network between the blood circulation around the ethmoidal bone and the brain.21,22 According to another view, SARS-CoV-2 has the ability to invade peripheral nerve terminals and slowly progresses through the synapse-related pathway to reach the CNS. In this context, it has been previously shown that when SARS-CoV and MERS-CoV were administered to mice via the nasal route, the virus reached the brain through the olfactory nerves and then affected different brain regions, including the thalamus and brainstem.20,21 Apart from the direct entry of the virus into the nervous system, COVID-19 infection may cause neurological problems due to widespread cardiopulmonary failure and metabolic abnormalities triggered by infection of SARS-CoV-2 or as a result of autoimmune mechanisms.23 In particular, by producing more inflammatory cytokines, the cytokine storm that takes place during the illness stimulates T cells, macrophages, and endothelial cells. Then, by triggering vascular leakage, complement activity, and the coagulation cascade, elevated interleukin (IL)-6 release damages neurons and the brain.24

    Turbulent in heart rate is a phenomena seen in electrocardiograms that reflects momentary hemodynamic disturbances resulting from ventricular premature beats and describes the short-term baroreflex-mediated variations in the duration of the sinus cycle that occur after spontaneous ventricular premature beats. When compared to the rate before the ventricular premature beats, the sinus rate in healthy persons temporarily increases, then decelerates, and finally returns to the basal rate.25 A momentary drop in blood pressure due to a ventricular premature beat activates baroreceptors, which in turn causes a decrease in the RR interval cycle lengths as determined by TO and an increase in heart rate brought on by vagal inhibition. Meanwhile, the ANS’s sympathetic arc is stimulated by temporary relative hypotension.26 Vascular resistance and systolic blood pressure gradually rise due to increased sympathetic activity. As a result, vagal activity rises once again and cycle durations are extended which is indicated as TS.26–29 Heart rate turbulence, therefore, requires a robust interaction of both the vagal and sympathetic systems. A change in either of these systems can result in the absence of normal heart rate turbulence.29 Some recent studies have shown that the HRT evaluation results performed by evaluating atrial premature beats in patients with supraventricular premature beats indicate blunted HRT and that these patients are prone to atrial fibrillation.7,8

    A triggering focal activator and alterations in the atrial electrophysiologic characteristics that might sustain AF are both components of the pathophysiology of AF.30 According to experimental research, parasympathetic activation significantly reduces the atrial effective refractory period, which makes it easier for AF to start and continue.31,32 Premature beats are necessary in HR turbulence analysis, which examines changes in autonomic control over time. Therefore, by examining the changes in the HR turbulence measurements before the onset of AF, we attempted to test the hypothesis that modified vagal reactions to atrial premature beats might start before the onset of paroxysmal AF.7 A brief initial acceleration followed by a lengthier period of sinus rhythm slowdown characterizes the considered normal HRT response. This results in a negative TO in individuals with an intact HRT response and a higher TS response than those with an impaired HRT response. Therefore, both TO and TS parameters are considered to be related to the balanced functioning of the ANS and baroreflex sensitivity.33

    If symptoms and signs persist for more than 12 weeks after COVID-19 infection and other causes are excluded, it is considered Long-COVID-19 syndrome.9 Although the pathogenesis of this period is not clear, persistent hyperinflammatory process, ongoing viral activity within the viral reservoir of the host, inadequate antibody response and enduring effects of tissue tropism are held responsible for the Long-COVID-19 syndrome. However, the presence and extent of organ damage, the variability in the time required for recovery of all organ systems, the severe acute disease period, intensive care syndromes, complications related to COVID-19 in the acute period, and the side effects of drugs used during the acute disease process are also factors.34 Long-COVID syndrome, the incidence of which varies between 10% and 60%, may include symptoms that concern many systems, such as fatigue, chest pain, shortness of breath, cough, decreased exercise intolerance, headache, and loss of taste and smell. In addition, palpitations, joint pain, muscle pain and weakness, insomnia, diarrhea, rash or hair loss, issues with balance and movement, issues with memory and focus, and cognitive problems, including deteriorating quality of life, can be observed.35 In COVID-19 individuals, sinus tachycardia is often the most prevalent arrhythmia.36 Nonetheless, the most likely pathogenic arrhythmias are ventricular tachycardia, atrial fibrillation, or atrial flutter.37,38 The pathophysiology of COVID-19-associated AF is not well understood, but some hypotheses that have been proposed include electrolyte and acid-base balance abnormalities that may occur in the acute phase of severe disease, increased adrenergic activation, decreased angiotensin-converting enzyme 2 (ACE2) receptor count, sialic acid-spike protein and CD147 interaction, and myocardial and endothelial damage caused by inflammatory cytokine storm.39 In the initial phase of severe COVID-19 illness, hypoxia and deficiencies in electrolytes have been commonly documented. These conditions are believed to lead to the emergence of abrupt arrhythmias.40 An arrhythmia occurring at this stage may persist in the post-COVID period and cause a feeling of palpitations. Several survey-based studies have reported that patients who had COVID-19 reported more complaints of palpitations than patients who had never had COVID-19.36,41 However, patients’ explanations of palpitations are subjective interpretations, differ based on how the questionnaire is designed, and are not supported by concrete electrocardiographic data. A recent study based on an online survey, in which no concrete electrocardiographic data were documented, suggested that the cause of these complaints reported by the participants was postural orthostatic tachycardia syndrome.36 As of today, objective ECG and Holter-ECG data are needed to show that subjective palpitation complaints described in the post-COVID-19 period indicate specific cardiac arrhythmias. A recently published study analyzing HRT of ventricular extrasystoles suggests that individuals who have recovered from severe COVID-19 may be more likely to develop malignant ventricular arrhythmia than individuals who have never had severe COVID-19.42 Another recent study examining the effects of the post-COVID-19 period on HRT reported that HRT values were blunted in the group with positive current COVID-19 test results compared to the group with negative test results.43 Both studies were based on ventricular extrasystoles. The current study analyzes the HRT of supraventricular ectopic beats and whether individuals who have recovered from COVID-19 are predisposed to developing AF in the future.

    Compared to the control group and other recovered COVID-19 subgroups, the recovered severe COVID-19 subgroup in the current investigation had a substantially lower atrial HRT Onset value and a significantly greater prevalence of aberrant atrial HRT Onset (Table 2) (Figure 3). Correlation analyzes revealed that abnormal atrial HRT Onset values were associated with the severity of recovered COVID-19 and chest severity score, but not with the time after recovery or history of positive PCR test results (Table 3) (Figures 4 and 5). Presence of abnormal atrial HRT onset using regression analyses and the atrial HRT Onset value was independently predicted by smoking, HT, recovery from severe COVID-19, and the chest CT severity level of recovered COVID-19 participants. The fact that atrial HRT values in group IV were blunted compared to other groups can be linked to the invasive and medicinal procedures used in the treatment plan for individuals with severe COVID-19 (the use of agents known to have harmful effects on the immune system and myocardium, such as steroids, in high doses eg, Prednisolone 1 gram/day, intubation), prolonged profound hypoxia, as well as to the being infected with high doses of SARS-CoV-2, which is believed to harm the ANS when a patient is unwell. The findings suggest that those have recovered from severe COVID-19 are more likely than those who have not had severe COVID-19 to have blunted atrial HRT and, thus, a greater risk of developing AF.

    This investigation demonstrates that the HRT Onset responses to atrial premature impulses are blunted in recovered severe COVID-19 individuals compared with the other groups. ANS dysfunction may be brought on by the neurological, cardiac, and systemic symptoms of COVID-19 and the medications used to treat it, particularly corticosteroids and an ANS dysfunction may result in deviant autonomic reflexes, such as the temporary augmentation of vagal outflow that lessens heart rate variations in response to premature impulses, which explains these deviating HRT reactions to atrial premature beats.

    There is a significant increase in the applications of recovered from COVID-19 individuals to psychiatric outpatient clinics with complaints of palpitations. In recently published articles on this subject, a parallel treatment protocol is introduced to the daily routine when palpitations are related to sadness, anxiety, or panic disorder.44–48 The COVID-19 process, medical and invasive treatment processes may lead to irreversible damage to the heart muscle itself, internal conduction pathways or ANS. It should be kept in mind any potential harm to the heart’s tissue, the ANS, or the intrinsic conduction pathways may cause supraventricular arrhythmia and therefore palpitation.

    Limitations

    Limitations of the study include the long-term effects of COVID-19 vaccines on the myocardium are not yet known and these vaccines were ignored when evaluating the cases, as well as the retrospective nature of the study.

    Conclusion

    This study demonstrated that abnormal atrial HRT independently was associated with recovering from severe COVID-19. In this context, this research offers verifiable proof of persistent palpitation complaints months after COVID-19 treatment, and it recommends a thorough 24-hour ECG-Holter examination for individuals who have recovered from severe COVID-19 to identify abnormal atrial HRT presence early and prevent AF.

    Ethical Approval

    The ethical approval was taken from the Ethics Committee of T.C. Fırat University (2021/12–45).

    Funding

    This article’s research, authoring, and/or publishing were all done without any financial assistance to the author/authors.

    Disclosure

    Regarding the research, authorship, and/or publishing of this paper, the authors have no possible conflicts of interest to report.

    References

    1. Demirhan R, Çimenoğlu B, Yılmaz E. The effects of hospital organization on treatment during COVID-19 pandemic. South Clin Ist Euras. 2020;31:89–95 doi:10.147/scie.2020.32154

    2. Wang D, Hu B, Hu C, et al. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus-infected pneumonia in Wuhan, China. JAMA. 2020;323(11):1061–1069. doi:10.1001/jama.2020.1585

    3. Yenerçağ M, Arslan U, Şeker OO, et al. Evaluation of P-wave dispersion in patients with newly diagnosed coronavirus disease 2019. J Cardiovasc Med. 2021;22(3):197–203. doi:10.2459/JCM.0000000000001135

    4. Yıldırım Y, Karaca O, Yılmaz FK, Güneş HM, Çakal B. Fragmented QRS on surface electrocardiography as a predictor of cardiac mortality in patients with SARS-CoV-2 infection. J Electrocardiol. 2021;66:108–112. doi:10.1016/j.jelectrocard.2021.03.001

    5. Bauer A, Malik M, Schmidt G, et al. Heart rate turbulence: standards of measurement, physiological interpretation, and clinical use: international society for holter and noninvasive electrophysiology consensus. J Am Coll Cardiol. 2008;52(17):1353–1365. doi:10.1016/j.jacc.2008.07.041

    6. Barthel P, Schneider R, Bauer A, et al. Risk stratification after acute myocardial infarction by heart rate turbulence. Circulation. 2003;108(10):1221–1226. doi:10.1161/01.CIR.0000088783.34082.89

    7. Vikman S, Lindgren K, Mäkikallio TH, Yli-Mäyry S, Airaksinen KJ, Huikuri HV. Heart rate turbulence after atrial premature beats before spontaneous onset of atrial fibrillation. J Am Coll Cardiol. 2005;45(2):278–284. doi:10.1016/j.jacc.2004.10.033

    8. Qu XF, Liu L, Guo XN, Piao JY, Gao GY, Huang YL. Investigation of heart rate turbulence after atrial premature beats before onset of paroxysmal atrial fibrillation. Zhonghua Yi Xue Za Zhi. 2007;87(40):2840–2842.

    9. Abdel-Gawad M, Zaghloul MS, Abd-Elsalam S, et al. Post-COVID-19 syndrome clinical manifestations: a systematic review. Antiinflamm Antiallergy Agents Med Chem. 2022;21(2):115–120. doi:10.2174/1871523021666220328115818

    10. Carfì A, Bernabei R, Landi F. Persistent symptoms in patients after acute COVID-19. JAMA. 2020;324:603–635.

    11. Tenforde MW, Kim SS, Lindsell CJ, et al. Symptom duration and risk factors for delayed return to usual health among outpatients with COVID-19 in a multistate health care systems network—United States. 2020. MMWR Morb Mortal Wkly. 2020;69(30):993–998. doi:10.15585/mmwr.mm6930e1

    12. Mallick D, Goyal L, Chourasia P, Zapata MR, Yashi K, Surani S. COVID-19 induced postural orthostatic tachycardia syndrome (POTS): a review. Cureus. 2023;15(3):e36955. doi:10.7759/cureus.36955

    13. Singh AK, Gupta R, Misra A. Comorbidities in COVID-19: outcomes in hypertensive cohort and controversies with renin angiotensin system blockers. Diabetes Metab Syndr. 2020;14(4):283–287. doi:10.1016/j.dsx.2020.03.016

    14. Verdecchia P, Cavallini C, Spanevello A, Angeli F. The pivotal link between ACE2 deficiency and SARS-CoV-2 infection. Eur J Intern Med. 2020;76:14–20. doi:10.1016/j.ejim.2020.04.037

    15. Bhatraju PK, Ghassemieh BJ, Nichols M, et al. COVID-19 in critically ill patients in the Seattle region – case series. N Engl J Med. 2020;382(21):2012–2022. doi:10.1056/NEJMoa2004500

    16. Oudit GY, Kassiri Z, Jiang C, et al. SARS coronavirus modulation of myocardial ACE2 expression and inflammation in patients with SARS. Eur J Clin Invest. 2009;39(7):618–625. doi:10.1111/j.1365-2362.2009.02153.x

    17. Ding Y, He L, Zhang Q, et al. Organ distribution of severe acute respiratory syndrome (SARS) associated coronavirus (SARS-CoV) in SARS patients: implications for pathogenesis and virus transmission pathways. J Pathol. 2004;203(2):622–630. doi:10.1002/path.1560

    18. Yashavantha Rao HC, Jayabaskaran C. The emergence of a novel coronavirus (SARS-CoV-2) disease and their neuroinvasive propensity may affect in COVID-19 patients. J Med Virol. 2020;92(7):786–790. doi:10.1002/jmv.25918

    19. Natoli S, Oliveira V, Calabresi P, Maia LF, Pisani A. Does SARS-Cov-2 invade the brain? Translational lessons from animal models. Eur J Neurol. 2020;27(9):1764–1773. doi:10.1111/ene.14277

    20. Li YC, Bai WZ, Hashikawa T. The neuroinvasive potential of SARS-CoV2 may play a role in the respiratory failure of COVID-19 patients. J Med Virol. 2020;92(6):552–555. doi:10.1002/jmv.25728

    21. Das G, Mukherjee N, Ghosh S. Neurological Insights of COVID-19 Pandemic. ACS Chem Neurosci. 2020;11(9):1206–1209. doi:10.1021/acschemneuro.0c00201

    22. Wu Y, Xu X, Chen Z, et al. Nervous system involvement after infection with COVID-19 and other coronaviruses. Brain Behav Immun. 2020;87:18–22. doi:10.1016/j.bbi.2020.03.031

    23. Berger JR. COVID-19 and the nervous system. J Neurovirol. 2020;26(2):143–148. doi:10.1007/s13365-020-00840-5

    24. Ahmad I, Rathore FA. Neurological manifestations and complications of COVID-19: a Literature Review. J Clin Neurosci. 2020;77:8–12. doi:10.1016/j.jocn.2020.05.017

    25. Schmidt G, Malik M, Barthel P, et al. Heart-rate turbulence after ventricular premature beats as a predictor of mortality after acute myocardial infarction. Lancet. 1999;353(9162):1390–1396. doi:10.1016/S0140-6736(98)08428-1

    26. Segerson NM, Wasmund SL, Abedin M, et al. Heart rate turbulence parameters correlate with post-premature ventricular contraction changes in muscle sympathetic activity. Hear Rhythm. 2007;4(3):284–289. doi:10.1016/j.hrthm.2006.10.020

    27. Berkowitsch A, Zareba W, Neumann T, et al. Risk stratification using heart rate turbulence and ventricular arrhythmia in MADIT II: usefulness and limitations of a 10-minute holter recording. Ann Noninvasive Electrocardiol. 2004;9(3):270–279. doi:10.1111/j.1542-474X.2004.93600.x

    28. Zuern CS, Barthel P, Bauer A. Heart rate turbulence as risk-predictor after myocardial infarction. Front Physiol. 2011;2:99. doi:10.3389/fphys.2011.00099

    29. Wichterle D, Melenovsky V, Simek J, Malik J, Malik M. Hemodynamics and autonomic control of heart rate turbulence. J Cardiovasc Electrophysiol. 2006;17(3):286–291. doi:10.1111/j.1540-8167.2005.00330.x

    30. Markides V, Schilling RJ. Atrial fibrillation: classification, pathophysiology, mechanisms and drug treatment. Heart. 2003;89(8):939–943. doi:10.1136/heart.89.8.939

    31. Liu L, Nattel S. Differing sympathetic and vagal effects on atrial fibrillation in dogs: role of refractoriness heterogeneity. Am J Physiol. 1997;273:805–816 doi:10.1152/ajpheart.1997.273.2.H805

    32. Geddes LA, Hinds M, Babbs CF, et al. Maintenance of atrial fibrillation in anesthetized and unanesthetized sheep using cholinergic drive. Pacing Clin Electrophysiol. 1996;19(2):165–175. doi:10.1111/j.1540-8159.1996.tb03308.x

    33. Iwasaki M, Yuasa F, Yuyama R, et al. Correla-tion of heart rate turbulence with sympathovagal balance in patients with acute myocardial infarction. Clin Exp Hypertens. 2005;27(2–3):251–257. doi:10.1081/CEH-48872

    34. Nikki N. Long covid: how to define it and how to manage it. BMJ. 2020;370:m3489. doi:10.1136/bmj.m3489

    35. Sudre CH, Murray B, Varsavsky T, et al. Attributes and predictors of Long-COVID: analysis of COVID cases and their symptoms collected by the covid symptoms study app. medRxiv. 2020. doi:10.1101/2020.10.19.20214494

    36. Davis HE, Assaf GS, McCorkell L, et al. Characterizing long COVID in an international cohort: 7 months of symptoms and their impact. EClin Med. 2021;38:101019. doi:10.1016/j.eclinm.2021.101019

    37. Ingul CB, Grimsmo J, Mecinaj A, et al. Cardiac dysfunction and arrhythmias 3 months after hospitalization for COVID-19. J Am Heart Assoc. 2022;11(3):e023473. doi:10.1161/JAHA.121.023473

    38. Spinoni EG, Mennuni M, Rognoni A, et al. Contribution of atrial fibrillation to in-hospital mortality in patients with COVID-19. Circ Arrhythm Electrophysiol. 2021;14(2):e009375. doi:10.1161/CIRCEP.120.009375

    39. Gawałko M, Kapłon-Cieślicka A, Hohl M, Dobrev D, Linz D. COVID-19 associated atrial fibrillation: incidence, putative mechanisms and potential clinical implications. Int J Cardiol Heart Vasc. 2020;30:100631. doi:10.1016/j.ijcha.2020.100631

    40. Guo T, Fan Y, Chen M, et al. Cardiovascular Implications of fatal outcomes of patients with coronavirus disease 2019 (COVID-19). JAMA Cardiol. 2020;5(7):1–8. doi:10.1001/jamacardio.2020.1017

    41. Huang C, Huang L, Wang Y, et al. 6-month consequences of COVID-19 in patients discharged from hospital: a cohort study. Lancet. 2023;401(10393):e21–e33. doi:10.1016/S0140-6736(23)00810-3

    42. Yılmaz M, Mirzaoğlu Ç. Retrospective cohort study: severe COVID-19 leads to permanent blunted heart rate turbulence. Diagnostics. 2025;15(5):621. doi:10.3390/diagnostics15050621

    43. Taş S, Taş Ü. Effects of COVID-19 on the autonomic cardiovascular system: heart rate variability and turbulence in recovered patients. Tex Heart Inst J. 2023;50(4):e227952. doi:10.14503/THIJ-22-7952

    44. Huang B, Yan H, Hu L, et al. The contribution of psychological distress to resting palpitations in patients who recovered from severe COVID-19. Int J Gen Med. 2021;14:9371–9378. doi:10.2147/IJGM.S334715

    45. Keshtkar A, Bahrami B. Relationship between COVID-19 anxiety and hypochondriasis in retirees aged 60 to 70 years in Shiraz. J Environ Treat Tech. 2021;9(4):737–740. doi:10.47277/JETT/9(4)739

    46. Kasi LS, Moorthy B. A case report on care-seeking type illness anxiety disorder after COVID-19 infection. Case Rep Psychiatry. 2023;2023:3003499. doi:10.1155/2023/3003499

    47. Kumar A, Cohen C. Post-COVID-19 panic disorder in older adults: two case reports. Am J Geriatric Psychiatry. 2021;29(4):58–59. doi:10.1016/j.jagp.2021.01.050

    48. Javelot H, Weiner L. Panic and pandemic: narrative review of the literatüre on the links and risks of panic disorder as a consequence of the SARS-CoV-2 pandemic. Encephale. 2021;47(1):38–42. doi:10.1016/j.encep.2020.08.001

    Continue Reading

  • Sugemalimab/Chemo Shows Durable Survival Benefit in First-Line Metastatic NSCLC

    Sugemalimab/Chemo Shows Durable Survival Benefit in First-Line Metastatic NSCLC

    Lung cancer: © Crystal Light – stock.adobe.com

    New, long-term follow-up data from the phase 3 GEMSTONE-302 trial (NCT03789604) confirm that the addition of sugemalimab (Cejemli) to first-line platinum-based chemotherapy provides a significant and durable overall survival (OS) benefit for patients with metastatic non–small cell lung cancer (NSCLC) who lack common genomic alterations. The 4-year follow-up, published in The Lancet Oncology, demonstrates that this combination therapy doubles the 4-year OS rate compared to chemotherapy alone, establishing it as a compelling standard of care option.1,2

    The multicenter, double-blind study enrolled 479 patients with metastatic squamous or nonsquamous NSCLC without known sensitizing EGFR, ALK, ROS1, or RET genomic alterations. Patients were randomized in a 2:1 ratio to receive either sugemalimab plus platinum-based chemotherapy (n = 320) or a placebo plus chemotherapy (n = 159). The primary analysis previously demonstrated significant improvements in both progression-free survival (PFS) and OS. This latest report, with a median follow-up of over 43 months, reinforces those initial findings with extended survival data.

    At the data cutoff in May 2023, the median OS for the sugemalimab group was 25.2 months (95% CI, 20.1–30.2), a substantial improvement over the 16.9 months (95% CI, 12.8–20.7) observed in the control group. This translated to a hazard ratio (HR) of 0.68 (95% CI, 0.54–0.85), indicating a 32% reduction in the risk of death. The survival benefit was particularly striking at the 4-year mark, with an OS rate of 32.1% in the sugemalimab arm compared with just 17.3% in the placebo arm. This long-term benefit was observed across different histological subtypes, with 4-year OS rates of 27.6% vs 11.7% in squamous NSCLC and 35.5% vs 20.2% in nonsquamous NSCLC.

    The initial benefit in PFS also remained robust over the extended follow-up. The median PFS was 9.0 months (95% CI, 7.4–10.9 months) with the addition of sugemalimab, a significant increase from the 4.9 months (95% CI, 4.8–5.2 months) in the control group (HR, 0.49; 95% CI, 0.39–0.60). This earlier finding highlighted the combination’s ability to delay disease progression effectively.

    In terms of safety, the long-term data from the GEMSTONE-302 trial did not introduce any new safety signals. The rates of treatment-related grade 3 or 4 adverse events were similar between the sugemalimab and control groups (56% vs 57%). The most common high-grade adverse events included decreased neutrophils (33% vs 33%), decreased white blood cells (15% vs 17%), and anemia (14% vs 11%). The incidence of treatment-related serious adverse events was 26% in the sugemalimab group and 20% in the control group. No new treatment-related deaths were reported since the previous interim analysis, which showed a treatment-related death rate of 3% versus 1%.

    The extended outcomes from the GEMSTONE-302 trial underscore the long-term efficacy and manageable safety profile of sugemalimab in combination with platinum-based chemotherapy.

    “These results underscore the efficacy of sugemalimab plus platinum-based chemotherapy as a standard first-line treatment option for both squamous and nonsquamous metastatic NSCLC while maintaining a manageable safety profile,” the study authors wrote. Findings support the continued use of this regimen as a potent first-line treatment for patients with metastatic NSCLC who do not have common driver mutations, offering clinicians a valuable tool to improve patient outcomes and survival duration.

    REFERENCES:
    1. Zhou C, Wang Z, Sun M, et al. Sugemalimab versus placebo, in combination with platinum-based chemotherapy, as first-line treatment of metastatic non-small-cell lung cancer (GEMSTONE-302): 4-year outcomes from a double-blind, randomised, phase 3 trial. Lancet Oncol. 2025 Jul;26(7):887-897. doi: 10.1016/S1470-2045(25)00198-6. Epub 2025 Jun 13.
    2. Stenger M. Extended Outcomes With Addition of Sugemalimab to First-Line Chemotherapy in Metastatic NSCLC. The ASCO Post. July 29, 2025. Accessed August 8, 2025. https://tinyurl.com/22e4txea

    Continue Reading

  • Micron raises forecasts as AI boosts memory chip demand – Reuters

    1. Micron raises forecasts as AI boosts memory chip demand  Reuters
    2. Micron’s Future: Growth or Bubble?  timothysykes.com
    3. Micron Updates Fourth Quarter Fiscal 2025 Guidance  GlobeNewswire
    4. Micron at KeyBanc Forum: AI and Memory Tech Drive Growth  Investing.com
    5. Micron stock enjoys large pre-market jump after quarterly results forecast raised  WSTM

    Continue Reading