Study Links Aggressive Prostate Cancer to Obesity


A new study from the Institut national de la recherche scientifique (INRS) shows that abdominal obesity appears to be associated with a greater risk of developing aggressive prostate cancer. The study was led by INRS Professor Marie-Élise Parent and was published in the journal Cancer Causes & Control.

Over the years, several studies have shown that obesity is a major risk factor for prostate cancer. To further explore the link between disease incidence and body mass, the research team studied data from a survey conducted in Montréal between 2005 and 2012. Researchers observed that abdominal obesity was associated with an increased risk of aggressive cancer.

“Pinpointing the risk factors for aggressive cancer is a big step forward in health research because it’s the hardest to treat,” said Prof. Parent. “This data creates an opportunity to work preventively, by monitoring men with this risk factor more closely,” she added.

The actual distribution of body fat appears to be a significant factor in the development of the disease: the impact on a person’s health can vary depending on whether the fat is concentrated around the abdomen or distributed throughout the body.

According to Éric Vallières, a Université de Montréal student conducting his doctoral research at INRS and the study’s main author, “Abdominal obesity causes hormonal and metabolic variations that can promote the growth of hormone-dependent cancer cells. Abdominal obesity is believed to be associated with a decrease in testosterone, as well as a state of chronic inflammation linked to the development of aggressive tumors.”

General obesity did not show the same correlation as abdominal fat. This may result from a detection bias and possible biological effects. “In obese people, the protein used to detect prostate cancer at an early stage, prostate-specific antigen (PSA), is diluted in the blood,” Mr. Vallières says. “This hemodilution makes cancer more difficult to detect.” 

The research team believes that studies on the timing of obesity exposure over a lifetime should be prioritized, and that a more in-depth analysis of body fat distribution could provide greater insight into the risks of developing prostate cancer.

Obesity associated with poorer mental health, especially in women


stress
Credit: Pixabay/CC0 Public Domain

A trio of mental and physical health researchers with University College Cork’s School of Public Health, has found evidence of poorer mental health in middle-aged to older people with obesity, independent of disease and lifestyle factors.

In their paper published on the open-access site PLOS ONE, Caoimhe Lonergan, Sean Millar, and Zubair Kabi, describe how they analyzed health data for more than 1,800 adult volunteers comparing BMI scores and mental health scores.

Prior research and anecdotal evidence have suggested a link between obesity and depression, but few studies have looked at the connection between the two using hard evidence from older people. For this new study, the research trio asked volunteers at a primary care center to participate in an obesity study.

As part of the study, each of the 1,821 volunteers (ages 46 to 73) gave permission and access to their medical records, and they also fasted overnight before providing blood samples, which were tested for glycated hemoglobin and glucose levels. Each was also measured for weight, height, and the circumference of their waist, which was used to calculate BMI. Each volunteer also filled out forms that described their lifestyle, demographics, and other health or disease factors.

In analyzing the data, and factoring out lifestyle factors, the researchers found what they describe as an association between BMI/body measurements consistent with obesity and depression along with feelings of low well-being. They noted that such relationships were more common for the women in the study than for the men. They also noted that their findings were consistent with those found in other similar research efforts.

The research team suggests that poorer mental health in obese older people is likely tied to social and physical factors, noting that there is social stigma, prejudice and sometimes discrimination associated with people suffering from obesity. They noted also that a number of studies have shown that there are many health problems associated with obesity, from joint and back pain to cardiovascular disease to fibromyalgia.

They suggest the combination of frustrations faced by obese people likely contributes to poor mental health. They conclude by suggesting that targeted interventions by health professionals should include weight management assistance.

Obesity disrupts mitochondria, reduces fat-burning


At a Glance

  • Scientists identified a protein that triggers changes in mitochondria that lead to reduced fat-burning to promote weight gain in mice fed a high-fat diet.
  • The findings may point to potential new avenues for treating or preventing obesity in people.

Mitochondria surrounding lipid droplets appear brighter in right panel.

A high-fat diet caused mitochondria (purple) in white fat cells to become fragmented and less effective at burning energy (left panel). In mice that lacked RalA, the absence of RalA prevented this fragmentation (right panel) and preserved the energy production of mitochondria. The large lipid (fat) droplets in fat cells appear green.

Adipose tissue, or body fat, plays a key role in maintaining our health. It helps to store and supply energy, regulate body temperature, and send hormone signals that affect many body functions. But when a person develops obesity, it leads to expansion of a type of fat called white adipose tissue, along with increased inflammation and metabolic changes.

Mitochondria, the energy-generating structures found within cells, are dynamic—that is, they can fuse, change shape, and divide. These changes affect how much energy mitochondria can burn. Some studies have found that obesity can alter these dynamics and cause mitochondria to fragment, making it more difficult for fat cells to burn energy. This might help explain why it can be hard for people with obesity to lose weight. The breakdown of mitochondria has also been tied to insulin resistance in obesity. And insulin resistance is associated with diabetes and other metabolic conditions. But the underlying connections between obesity, mitochondria, and white fat have been unclear.

A research team led by Dr. Alan Saltiel at the University of California, San Diego, had previously shown that a protein called RalA could be activated by insulin in fat cells and promote glucose uptake by brown fat. The team suspected that study of RalA might also give insights into the mitochondrial changes linked to obesity.

To investigate, the researchers fed mice a high-fat diet (about 60% fat) for 8 to 12 weeks. They then examined its effects on RalA and the mitochondria in fat cells. Results were reported on January 29, 2024, in Nature Metabolism.

The researchers found that the high-fat diet caused the mitochondria in white fat tissue to divide into many smaller pieces. As a result, the mitochondria became less effective at burning energy. The high-fat diet also boosted levels of RalA in white fat. These changes weren’t seen in brown fat tissue. The results hinted that higher RalA activity might be the culprit behind many metabolic problems seen in obesity.

To gain insights into the effects of RalA activation, the team looked at what happens in the protein’s absence. They created genetically altered mice that lacked the RalA-producing gene in fat tissues. When given a high-fat diet, mice without RalA in their white fat tissue were protected against diet-induced weight gain and obesity. They had additional metabolic improvements as well. These included better liver function and glucose tolerance, and energy expenditure similar to mice fed a regular diet. Absence of RalA also prevented fragmentation of mitochondria in mice fed a high-fat-diet, thereby protecting mitochondria’s fat-burning functions.

Further analysis showed how RalA activity leads to changes in mitochondria dynamics. The researchers found the same mechanisms in white fat from people. They also found that the activity of a key protein in the process was associated with human obesity. More study will be needed to understand how a high-fat diet raises levels of RalA in white fat in the first place.

“In essence, chronic activation of RalA appears to play a critical role in suppressing energy expenditure in obese adipose tissue,” Saltiel says. “By understanding this mechanism, we’re one step closer to developing targeted therapies that could address weight gain and associated metabolic dysfunctions by increasing fat burning.”

Daily servings of kimchi may reduce obesity, but researchers urge moderation


Key takeaways:

  • Three or more daily servings of cabbage kimchi was associated with 10% reduced odds of obesity in men.
  • Excessive consumption of kimchi could still lead to obesity, researchers warned.

Daily consumption of one to three servings of kimchi was associated with a decreased prevalence of obesity in men, according to a study in BMJ Open.

Hyien Jung, from the department of food and nutrition at Chung Ang University in South Korea, and colleagues explained that kimchi — a dish made of fermented salt and vegetables — has previously shown anti-obesity effects in animal studies because of its lactic acid bacteria.

PC0224Jung_Graphic_01_WEB
Three or more daily servings of cabbage kimchi was associated with 10% reduced odds of obesity in men.

However, “there are currently a few epidemiology studies investigating the relationship between kimchi consumption and obesity in adults,” they wrote.

In the study, the researchers examined the diets of 115,726 Korean participants aged 40 to 69 years between 2004 and 2013. They were part of a prospective cohort study of environmental and genetic risk factors for common chronic diseases.

They found that in men, consumption of one to two servings (OR = 0.875; 95% CI, 0.808-0.947) and two to three servings (OR = 0.893; 95% CI, 0.817-0.978) of kimchi per day were associated with a lower prevalence of obesity vs. consumption of less than one serving per day.

Men who had three or more daily servings of a specific type of the dish — cabbage kimchi — had 10% reduced odds of obesity (OR = 0.904; 95% CI, 0.832-0.982) and abdominal obesity (OR = 0.903; 95% CI, 0.825-0.989) compared with those who had less than a serving per day.

Jung and colleagues added that there was a lower prevalence of obesity in both men (OR = 0.908; 95% CI, 0.842-0.979) and women (OR = 0.895; 95% CI, 0.855-0.938) who consumed less than the median daily amount of radish kimchi — 25 g for men and 10.7 g for woman. Also, conversely, consumption of radish kimchi exceeding the median daily amount was associated with a lower prevalence of abdominal obesity in men (OR = 0.915; 95% CI, 0.84-0.996) and women (OR = 0.889; 95% CI, 0.842-0.939) vs. nonconsumers.

There were some limitations in the study. For example, its cross-sectional design impacted the researchers’ ability to make a casual inference.

“Thus, a longitudinal study is necessary to better understand the impact of kimchi on obesity,” they wrote. “Furthermore, this finding cannot be generalized due to the study’s focus on Korean participants.”

Jung and colleagues pointed out that increased total kimchi consumption was tied to higher energy, carbohydrate, protein, sodium and fat intake, which “might lead to increased weight.”

“Since all results showed a ‘J-shaped’ association, excessive consumption suggests the potential for an increase in obesity prevalence,” they concluded. “As kimchi is one of the major sources of sodium intake, a moderate amount of kimchi should be recommended for the health benefits of its other components.”

Obesity linked with higher risk of early sepsis-associated AKI in adults admitted to ICU


Key takeaways:

  • Overall, 33.8% of patients admitted to an ICU developed early sepsis-associated AKI.
  • Obesity correlated with a higher incidence of sepsis-associated AKI compared with normal weight.

Obesity may be linked to a higher risk of developing early sepsis-associated AKI in adults admitted to the ICU, according to a recently published study.

Researchers led by Yoon Hae Ahn, MD, of the Seoul National University Hospital, ran a nationwide prospective cohort trial of 4,041 patients aged 19 years or older admitted to 20 tertiary hospital ICUs in Korea from 2019 and 2021 to explore related clinical outcomes.

Ahn_Graphic
Data derived from Ahn YH, et al. JAMA Netw. Open. 2024;10.1001/jamanetworkopen.2023.54923.

“Obesity is rising in ICUs worldwide … Multiple studies have shown an association between obesity and the development of AKI in patients with critical illness,” Ahn and colleagues wrote. “Sepsis-associated AKI (SA-AKI) is associated with poor clinical outcomes, including a higher risk of in-hospital mortality, longer hospital stays and a greater chance of [chronic kidney disease] CKD.”

After excluding patients with preexisting stage 3A to 5 CKD from the study, the remaining patients were categorized by BMI.

The main outcome was the incidence of SA-AKI within 48 hours of ICU admission, and secondary outcomes were mortality and clinical recovery, defined as survival to discharge within 30 days.

Findings showed 33.8% of patients developed early SA-AKI. Obesity correlated with a higher SA-AKI incidence compared with normal weight, and obesity was tied to lower in-hospital mortality in patients without SA-AKI compared to those without obesity. Researchers categorized weight into four groups of BMI: underweight for a BMI less than 18.5 kg/m2, normal weight for 18.5 kg/m2 to 22.9 kg/m2, overweight between 23 kg/m2 and 24.9 kg/m2and obesity defined as 25 kg/m2or greater.

Researchers found no difference in mortality for patients with SA-AKI, adding that while patients with obesity and without SA-AKI had a greater likelihood of clinical recovery compared to those without obesity, it was less likely in patients with both conditions.

The results “highlight the need for future research on the mechanisms underlying the complex association among obesity [and] SA-AKI and clinical outcomes in patients with sepsis,” Ahn and colleagues wrote.

Tirzepatide Reduces 10-Year Atherosclerosis Risk Among Patients With Obesity


Atherosclerosis. Computer artwork of a narrowed artery, due to a cholesterol plaque.

Tirzepatide was linked to improved risk for atherosclerosis among patients with obesity.

Tirzepatide treatment is associated with a significant reduction in 10-year risk for atherosclerosis among individuals with obesity or overweight and without diabetes, according to study findings published in Diabetes, Obesity and Metabolism.

Findings from the phase 3, randomized clinical SURMOUNT-1 (ClinicalTrials.gov; Identifier: NCT04184622) trial supported the use of tripeptide for chronic weight management among patients with obesity. However, the effect of tirzepatide on long-term risk for atherosclerosis remains unknown.

To assess the relationship between tirzepatide and obesity-related cardiovascular complications, researchers conducted post-hoc analysis of the SURMOUNT-1 clinical trial using participant data from baseline up to week 72. Participants were randomized to receive either 5, 10, or 15 mg tirzepatide or placebo weekly as an adjunct to lifestyle intervention.

Inclusion criteria were adults with obesity or overweight without diabetes. The researchers excluded patients with a history of cardiovascular disease for this analysis.

Primary outcomes were changes in antihypertensive and antihyperlipidemic therapy, waist circumference, blood pressure, glycated hemoglobin, and fasting glucose. The researchers calculated the change in 10-year atherosclerosis risk using the American College of Cardiology and American Heart Association risk engine, which stratified risk scores according to:

  • Low-risk (<5%);
  • Borderline-risk (5-7.5%);
  • Intermediate-risk (7.5-20%); and,
  • High-risk (≥20%).

The study population included 2461 participants, of whom 622, 614, 616, and 609 were assigned to the placebo and tirzepatide 5 mg, 10 mg, and 15 mg groups, respectively. The mean age was 44.5 years (SD, 12.3) and the mean body mass index was 38.0 kg/m2. Among the study population, 68.4% were women and 70.3% were White.

At baseline, the proportions of participants with low-risk, borderline-risk, intermediate-risk, and high-risk atherosclerosis risk scores were 80.4%, 8.6%, 10.0%, and 1.0%, respectively. Baseline median 10-year risk scores ranged between 1.5% and 1.6% and did not vary between treatment groups.

The researchers noted significantly greater reductions in atherosclerosis risk among the treatment groups compared with placebo. At week 72, the relative changes in predicted 10-year atherosclerosis risk were -16.4% (tirzepatide 5 mg), -23.5% (tirzepatide 10 mg) and -22.4% (tirzepatide 15 mg) compared with 12.7% (placebo; P <.001).

Among the subset of participants with baseline intermediate- and high-risk scores, the relative changes in atherosclerosis risk scores at week 72 were -10.3% (tirzepatide 5 mg), -20.6% (tirzepatide 10 mg), and -16.1% (tirzepatide 15 mg) compared with 6.4% (placebo; P <.05).

Compared with placebo, participants treated with tirzepatide had significantly improved odds of achieving reduced atherosclerosis risk at week 24 (odds ratio [OR], 2.2; 95% CI, 1.6-3.0; P <.001) and week 72 (OR, 2.4; 95% CI, 1.7-3.5; P <.001). Similarly, tirzepatide-treated participants with intermediate- and high-risk scores at baseline had significantly increased odds of achieving reduced atherosclerosis risk at week 24 (OR, 2.8; 95% CI, 1.4-5.6; P =.003) and week 72 (OR, 2.9; 95% CI, 1.3-6.2; P =.008).

Study limitations include the fact that the atherosclerosis risk engine was not developed or validated among populations of patients with obesity or overweight exclusively.

“[T]reatment with tirzepatide significantly reduced the 10-year predicted risk of [atherosclerosis] compared with placebo in people with obesity or overweight but without diabetes,” the researchers wrote. “The absolute reduction in risk was greater for participants with higher [atherosclerosis] risk at baseline.”

The ‘Triple G’ Agonist for Obesity Management: 5 Things to Know


The complex pathophysiology of obesity requires a multidisciplinary approach that includes lifestyle and medical interventions for successful management. Anti-obesity medications (AOMs) have emerged as a powerful and life-changing tool for many individuals with obesity who are unable to sustain long-term weight loss through lifestyle changes alone. As with other chronic diseases such as hypertension and hyperlipidemia, the goal of decades of research has been to develop anti-obesity medications with long-term efficacy and safety. Recent groundbreaking findings from a phase 2 trial show immense potential for a new AOM.

Here are five things to know about the role of agonists in the management of obesity.

1. Gut hormone physiology informs the development of AOMs.

The three hormones associated with obesity or diabetes are glucagon-like peptide 1 (GLP-1), glucose-dependent insulinotropic peptide (GIP), and glucagon. GLP-1, a peptide released from the intestines in response to food ingestion, increases insulin production, reduces gut motility, and suppresses appetite. GIP is also an intestinal hormone that increases meal-stimulated insulin production and additionally facilitates lipolysis. Glucagon is known to increase hepatic glucose output but will also increase insulin secretion in the setting of hyperglycemia. Glucagon also promotes lipolysis.

Though these hormones are more commonly thought of as incretins, gut hormones that stimulate postprandial insulin secretion, their role in energy physiology is more diverse. Due to multiple mechanisms of action, incretins are increasingly referred to as nutrient-stimulated hormones (NuSH), a term which encompasses other peptides with therapeutic potential (eg, amylin, oxyntomodulin, peptide tyrosine tyrosine).

2. Studies have shown that NuSH therapies are highly effective AOMs.

In 2021 the US Food and Drug Administration approved subcutaneous semaglutide 2.4 mg, a GLP-1 receptor agonist, for the treatment of obesity. Clinical trials demonstrating an average weight loss of 15% in patients taking semaglutide ushered in a new era of AOMs associated with significant weight loss that not only improves disease activity but also has the potential to achieve diabetes remission. Recent findings from the OASIS I trial demonstrated an average weight loss of 15.1% from baseline in patients treated with oral semaglutide for 68 weeks. Medical societies, including the American Diabetes Association and the American Association for the Study of Liver Diseases, recommend 10%-15% weight loss to fully treat weight-related comorbidities like type 2 diabetes and nonalcoholic fatty liver disease. In 2022, tirzepatide, a dual GLP-1 and GIP receptor agonist, demonstrated an average weight loss of 22.5% in phase 3 of the SURMOUNT-1 trial for obesity — a weight loss approaching that of some bariatric surgeries.

3. Clinical trial data show that the novel triple agonist retatrutide induces significant weight loss.

Preclinical studies on the newest NuSH therapy, triple GLP-1–GIP–glucagon receptor agonist retatrutide showed predominant activity at the GIP receptor, with less GLP-1– and glucagon-receptor agonism than that of endogenous GLP-1 and GIP. Results from a phase 2 trial published in June 2023 showed a weight loss of 24% at 48 weeks in adults with obesity treated with retatrutide, which is the greatest weight loss reported in an obesity trial so far. Moreover, for the first time in obesity pharmacotherapy research, 100% of participants achieved clinically significant weight loss (defined as ≥ 5% of baseline weight).

4. Retatrutide may improve lipid metabolism.

In the phase 2 trial, retatrutide reduced low-density lipoprotein cholesterol (LDL-C) levels by approximately 20%. This degree of reduced plasma LDL-C is dramatic in weight loss studies. Typically, weight loss significantly reduces triglyceride levels, increases high-density lipoprotein cholesterol levels, and has a modest effect on LDL-C reduction of about 5%.

A 20% reduction in LDL-C with retatrutide is hypothesis-generating. Preclinical studies have shown glucagon to be an important regulator of proprotein convertase subtilisin/kexin type 9degradation, with the lack of glucagon resulting in increased PCSK9 levels, decreased LDL receptors, and increased plasma LDL; conversely, treatment with glucagon decreased plasma LDL.

5. The long-term safety of retatrutide still needs to be determined.

In the 48-week phase 2 trial, retatrutide was observed to have a side effect profile largely similar to other NuSH therapies (eg, semaglutide 2.4 mg, tirzepatide), with a predominance of gastrointestinal symptoms including nausea, diarrhea, vomiting, and constipation. However, side effects potentially unique to retatrutide also emerged. Cutaneous hyperesthesia and skin sensitivity were reported in 7% of participants in the retatrutide group vs 1% in the placebo group; none of these effects were associated with physical skin findings. Of note, 17 out of 198 (9%) participants in the retatrutide group developed cardiac arrhythmia vs two out of 70 (3%) in the placebo group. There was no consistent pattern of arrhythmia type (eg, supraventricular, ventricular) observed, and some of these events were reported as “palpitations” or “increased heart rate” without further detail. Phase 3 clinical trial data will provide further insight into the long-term safety of retatrutide.

Diabetes, Eating Disorders, and Obesity: Navigating the Complexities of Treatment


People with diabetes are at a higher risk of developing eating disorders. Things get even more complicated when obesity is involved. Here’s why compassionate care and specialized treatment are so important.

Sugar, cigarettes, weed, alcohol, cocaine, and then food. Monica Romano, a 55-year-old living in Bellingham, Washington, lists her “drugs” in order of addiction. 

Romano quit drugs like cocaine at the age of 30 but then developed another addiction.

“Food became my new drug,” she said. She described her first time binge eating feeling as natural as breathing. Now she identifies as a food addict, with obesity as one of the symptoms. 

Looking back, she realizes she had binge eating disorder – a chronic condition marked by compulsive overeating – but never discussed it with doctors. 

Due to her weight, family history, and diagnosis of metabolic syndrome, Romano worries about the ever-present threat of diabetes. Metabolic syndrome increases the risk for health conditions like diabetes, heart disease, and stroke. Many people with metabolic syndrome also have insulin resistance, which over time may lead to type 2 diabetes.

“I have luckily managed to escape it so far. But the threat is always at the back of my mind,” she said. 

Identifying and treating each of her conditions, while working to avoid a diabetes diagnosis, has been a long and difficult journey, fraught with stigma and challenges.

Three complex, intertwined conditions

Dealing with the ups and downs of one medical journey can be daunting. Dealing with three simultaneously can seem downright impossible. 

But that’s exactly where some people are, as they navigate complications and conflicting advice on living with and treating obesity, eating disorders, and diabetes. This is becoming a more prevalent issue as clinical obesity rates continue to rise – almost 41% of Americans live with obesity, according to the Centers for Disease Control and Prevention (CDC).
 
Clinical obesity is a disease defined by the CDC as someone who has a body mass index (BMI) of 30 or higher. It’s important to note that BMI doesn’t accurately measure body fat content; while it can be used as a screening tool, BMI is not a reliable measure for diagnosing obesity. 

Along with the rising obesity rates, diabetes cases have doubled in the last 20 years, with overweight and obesity being a major risk factor for type 2 diabetes. Even though not everyone with diabetes has excess weight, 80-90% of the type 2 diabetes population has overweight or obesity.

People with diabetes also have higher rates of eating disorders than the general public, with 20% experiencing an eating disorder at some point. Bulimia nervosa is a more common eating disorder in type 1 diabetes and binge eating without purging is more common with type 2 diabetes.

The need for awareness and understanding around treatment

People with obesity, diabetes, and eating disorders can experience dilemmas in treatment that contradict each other – what helps with one condition might fuel another. 

Dr. Ann Goebel-Fabbri is a clinical psychologist who works with people managing these conditions (sometimes all three at once). She recalled an instance of one patient with overweight and type 2 diabetes who sought treatment to stabilize her binge eating disorder. 

However, the recommended weight management meal plan they recommended to help with her eating disorder conflicted with the recommended treatment for diabetes and obesity. 

“The problem was her meal plan at the treatment center completely ignored that to improve health in diabetes, a modest amount of weight loss is actually indicated,” Goebel-Fabbri said. 

“So the question is, how do we approach those concepts in ways that allow enough complexity, variation in foods, enjoyable foods, and prevent concepts and feelings like deprivation, shame, and failure?” she said. 

Goebel-Fabbri added that treatment must involve an understanding that deprivation doesn’t work. Eliminating a whole food group, as is the case with very low-carb diets, intermittent fasting, and other trendy fad diets, is actually more likely to increase the likelihood of binging. She said that it also only focuses on short-term success, rather than a long-term plan. 

“We need to help people develop modest and realistic goals for weight loss,” she said.

Pursuing multi-faceted, meaningful treatment

Romano is getting treatment at a clinic specializing in obesity and is also in counseling to help her better understand the link between trauma, post-traumatic stress disorder, and eating disorders. 

“That was eye-opening because it explained why I felt compelled to binge as a response to others noticing my weight loss,” she said. “In some strange way losing weight became synonymous with losing control. I’m still grappling with that one – this hasn’t been an easy fix.” 

“Sure, weight loss drugs and even bariatric surgery, which I considered for a while, can help but they are tools. Eating disorders are multifaceted and without mindset work, we miss the complete picture,” Romano said. 

Alle C. Hall lives with prediabetes and has had binge eating disorder her whole life. At times, she’s also dealt with bulimia and exercise compulsion. 

Along the way, Hall said that some primary care doctors and OB-GYNs have impeded progress in getting treatment. Their weigh-ins and lectures about diet and exercise have led to sometimes days-long binging episodes, she said. 

Instead, she hopes digging into past trauma will get to the root cause and help with healing. For her, writing a book was a part of healing. She encourages others to pursue a project or creative endeavor as it could be a valuable part of the healing process. 

“There is no need to spend your time feeling like you have no willpower,” Hall said. “I know this to be true: get the help you need.”

A year in review: The field of obesity medicine in 20


In the landscape of health care, the year 2023 witnessed remarkable strides in the field of obesity medicine.

These were marked by groundbreaking developments in anti-obesity medications, enhancements in access to care and insurance coverage for obesity treatment and an increase in the number of health care providers expressing an interest in specializing in obesity medicine. These highlights show the ongoing commitment to addressing this complex disease, offering hope and opportunities for individuals with obesity while reshaping the approach of health care professionals in their pursuit of comprehensive and effective obesity management strategies.

Overweight_scale_tape_268233374
Notable developments in obesity medicine in 2023 include the increase in semaglutide use for weight loss and the FDA approval of Zepbound.

This article explores the advancements and trends that shaped the field of obesity medicine in 2023, shedding light on the transformative impact these developments have had on patient care and the role of health care providers in the field.

Developments in anti-obesity medications

In 2023, the realm of anti-obesity medications witnessed an era of innovation, with developments that offer new avenues for obesity treatment. These medications, often developed with a more nuanced understanding of obesity, aim to provide solutions that go beyond traditional approaches.

Among the breakthroughs is the emergence of medications that not only aid in weight loss but also address underlying factors contributing to obesity-related comorbidities. Researchers and pharmaceutical companies collaborated to refine existing medication formulations and introduce new medications that demonstrate enhanced efficacy and safety. In 2023, more notable developments include the increase in the use of semaglutide medications such as Ozempic and Wegovy (Novo Nordisk) for weight loss. We also saw other forms of weight loss medications emerge, with the FDA approval of Eli Lilly’s tirzepatide medication, Zepbound. In addition to the new medications,The New England Journal of Medicine released the results of the SELECT study, showing the positive impacts of semaglutide medications on patients with cardiovascular disease and obesity.

The evolving landscape of anti-obesity medications reflects a commitment to offering individuals diverse and personalized treatment options, marking a significant stride forward in the medical community’s efforts to combat obesity. As these pharmaceutical innovations continue to unfold, they hold the promise of reshaping the standard of obesity management, providing new hope for patient outcomes and long-term success.

Access to care

While pharmacotherapy treatment options were expanding, many patients were still left wondering if they would have access to obesity care. Historically, individuals with obesity faced barriers to accessing comprehensive obesity treatment, often encountering limited health insurance coverage or providers unwilling to discuss treatment options because of lack of knowledge, resources or even weight bias. A growing recognition of the profound health implications associated with obesity should prompt insurers and health care systems to reassess their policies, leading to a welcome expansion in coverage and options for obesity treatments. This shift would not only alleviate the financial burden on individuals seeking assistance but also empower health care providers to offer more diverse and tailored interventions, fostering a more holistic and patient-centric approach to obesity care.

Throughout the medical community, there was a surge in efforts to improve access to obesity care for people with obesity. Recognizing the urgent need for a multifaceted approach to obesity, various organizations and associations have worked diligently to enhance the availability and accessibility of obesity-related services. The AMA has dedicated a large portion of their work to advocating for proper insurance coverage for obesity treatment and improving the narrative surrounding obesity treatment. In addition, numerous advocacy efforts for obesity treatment emerged, with the introduction of the Treat and Reduce Obesity Act of 2023, aimed at allowing Medicare to cover anti-obesity medications.

The growing focus on accessibility not only makes it easier to intervene early and implement preventive measures but also tackles the disparities that might have previously discouraged individuals with obesity from seeking and receiving effective care. As these initiatives gain momentum, the health care landscape is evolving to ensure that obesity care is not only possible from a medical standpoint, but also readily accessible to those who need it most.

Increase in interest for obesity medicine

Although advocating for proper access and coverage for obesity treatment is vital to ensuring positive outcomes for individuals with obesity, it cannot be done without one very important population: health care providers. In 2023, there was an increase in the number of health care providers expressing an interest in specializing in obesity medicine. This surge can be attributed to a growing recognition of the role that obesity plays in overall health, as well as an increasing awareness of the complexities involved in obesity management. In 2023, the American Board of Obesity Medicine had a record of 1,889 physicians from the United States and Canada apply to take the American Board of Obesity Medicine (ABOM) exam, representing a variety of fields of medicine, from internal medicine to obstetrics/gynecology. The exam brought the number of ABOM diplomates from 5,881 to 6,729 and tripled the number of diplomates since 2017.

The heightened interest in specializing in obesity medicine has shown how medical practitioners are recognizing that obesity is intricately linked to a myriad of comorbidities such as diabetes, cardiovascular diseases and certain cancers. Consequently, health care providers are motivated to delve into the field of obesity medicine to acquire the necessary skills and knowledge to offer more effective treatments, thereby contributing to improved patient outcomes. This trend not only signifies a crucial step forward in addressing the disease of obesity but also reflects a commitment within the health care community to confront this complex health challenge head-on.

The year 2023 proved to be a crucial moment in the field of obesity medicine, witnessing advancements that have reshaped the landscape of obesity management. The developments in anti-obesity medications have opened new avenues for tailored and effective treatments, marking a significant stride forward in the fight against obesity. Access to care and insurance coverage continue to be barriers for patients, so advocacy efforts must continue. The increase in the number of health care providers expressing an interest in specializing in obesity medicine reflects a transformative shift in the perception of obesity as a chronic medical condition. Although there is still much work to be done on all fronts regarding obesity medicine, the collective efforts of 2023 have set the stage for continued progress and a more compassionate, informed and effective approach to obesity care in the years to come.

Obesity’s link to depression tied to both physical and social factors


New research out of the United Kingdom is further re-enforcing the connection between weight issues and mental health. While this isn’t the first study to establish a connection between obesity or high body mass index (BMI) and depression, a team from the University of Exeter set out to answer how exactly a high BMI leads to depressive symptoms. Study authors report that a combination of both physical and social factors are likely at play.

Studies have linked excess weight to an increased risk of heart attack, stroke, and diabetes for decades. More recently, however, new studies have established a connection between obesity and poor mental health. Study authors conducted this new research in an effort to find out whether the link between high BMI and depression is a psychosocial or a physical reaction.

Psychosocial pathways include the social stigma or shame that many may feel about being overweight around others in better shape. There is also a societal perception that everyone should be skinny or in shape. Meanwhile, physical pathways are metabolic conditions with a historic connection to a high BMI, such as type 2 diabetes or high blood pressure.

Regardless of disease, weight impacts mental health

The team analyzed genetic data on over 145,000 people originally collected for the U.K. BioBank project during this study. They also examined any available mental health information from participants as well. Study authors then analyzed genetic variants associated with higher BMI in conjunction with scores on a clinically-relevant mental health survey put together to assess depression, anxiety, and well-being levels.

Obesity and depression are both major global health challenges, and our study provides the most robust evidence to date that higher BMI causes depression. Understanding whether physical or social factors are responsible for this relationship can help inform effective strategies to improve mental health and wellbeing,” says co-lead study author Jess O’Loughlin from the University of Exeter Medical School in a release. “Our research suggests that being fatter leads to a higher risk of depression, regardless of the role of metabolic health. This suggests that both physical health and social factors, such as social stigma, both play a role in the relationship between obesity and depression.”

Finding a genetic link between weight and mental health

To determine which pathways specifically lead to depression among overweight individuals, researchers also analyzed two recently discovered genetic variants. The first of those variants promotes obesity, albeit a much more metabolically healthy version of obesity. So, people with that set of genes may be overweight, but they’re actually much less likely to develop high blood pressure or other conditions. The second set of genes causes a more traditional version of obesity that’s linked to various metabolic problems.

Interestingly, study authors noted very little difference between the two variants, indicating that both social and physical elements play into the obesity-depression relationship.

“This is a robust study, made possible by the quality of UK Biobank data. Our research adds to a body of evidence that being overweight causes depression. Finding ways to support people to lose weight could benefit their mental health as well as their physical health,” concludes co-lead study author Dr. Francesco Casanova.