Chapters
Transcript
DR. MARK DYLEWSKI: Good evening. My name is Dr. Mark Dylewski. I would like to welcome our distinguished speakers and all the attendees to the Miami Cancer Institute lung cancer conference. This is a collaborative effort between the Miami Cancer Institute and the Lynn Cancer Center.
This evening, we have a highly distinguished group of faculty speaking this evening. And for the sake of time, I'm going to limit the introductions to provide enough time for the speakers to cover their talks. So our first speaker this evening is Dr. John Roberts, who is the Chief of Thoracic Surgery at the Lynn Cancer Center.
And he will be speaking on minimally invasive surgery for the treatment of lung cancer. Welcome Dr. Roberts.
DR. JOHN ROBERTS: Thanks, Dr. Dylewski. I appreciate being here. I'm John Roberts. I'm the thoracic surgeon up at Boca Raton, which is the little brother North of the Cancer Institute. I'm delighted to be here. All right, so I want to talk about the minimally invasive surgery in the treatment of lung cancer.
Minimally invasive surgery first began some 25 years ago, and I've been doing thoracoscopic lung resections for 25 years and robotic surgery for approximately 10 years. During this talk, I wanted to try to describe what I think a good cancer operation means. I want to briefly cover staging criteria for lung cancer.
I want to discuss some of the limitations of radiologic staging for lung cancer. I'll discuss briefly some techniques for surgical staging, review the literature that compares SBRT and surgery, and then just briefly cover the incisions that we use.
All right, so why should we operate on someone for lung cancer when we've got safer options? You can see this is just a slide that briefly demonstrates the incidence of lung cancer worldwide. The US and Europe are amongst the highest, with some 30 to 40,000-- 40 cases per 1,000.
We've improved over time with the survival of non-small cell lung cancer. Five year survivals for the various classifications of stage one range from 68% to 92%. For stage two, 53 to 60%. And then stage threes begin to fall off. All of these numbers are about 20 percentage points better than we used to do 20 years or so ago.
All right, so there are several options for lung surgery. The standard is just a postal lateral thoracotomy. This was basically what I was trained primarily on. About 25 years ago, we began to use thoracoscopic surgery, which requires an access incision here and either two or three ports inferiorly.
And the latest and probably best iteration is robotic surgery. Again, requires an access incision here with somewhere between three and four additional ports for a total of five ports. A lot of studies have shown significant benefits as far as shorter recovery time after minimally invasive surgery.
And so if possible, thoracoscopic or robotic surgery should be the standard. We still occasionally have to make the larger incisions for larger tumors. All right, so-- to delve into why we consider surgery, certainly one of the techniques that's been a dramatic improvement in the treatment of lung cancer has been the development of SBRT.
There are few studies that show that SBRT in surgery might be equivalent. I'll lump these two together because they've actually been written together. There were two separate small prospective randomized trials, randomized patients between SBRT and surgery.
They were each supposed to get around 500 patients, but were closed early and together had a total of 60 patients. SBRT had about a 95% three year survival, and surgery a three year survival of 79%. Some of the problems with this was that it was a very small size study, obviously.
Did not reach the numbers that they wanted to get. And a lot of the operations were wedge resections, which we know is not an adequate operation in general.
In contrast, there's a large number of studies that show the superiority of surgery to SBRT with survival. None of these are randomized because there have been no other randomized trials done. But you can see that some of the numbers are very large. This particular study, this [INAUDIBLE] study, chose patients within the group that had been recommended to have resection and elected to have SBRT instead, and their survival was significantly less.
I won't spend all-- a lot of time on these numbers, other than just to point out that there are significant differences, and the average difference ranges between 15 to about a 30% improvement in survival with surgery. So that's why we still try to operate on patients if we can.
All right, so let's talk about staging. There's also some limitations with radiologic staging. And I just want to give as an example, a patient. This is-- VR is a 76-year-old female presented with a lung mass. Chest CT demonstrated Tucson meter mass in the left upper lobe.
She was referred to our multimodality clinic where the patient's surgeon, radiation oncologist, and medical oncologist and we obtained a PET scan. The PET scan was interpreted as showing a stage one cancer with no evidence of distant disease, period. She appeared to be healthy and we were inclined to surgery, but she was inclined to radiation.
We were able to convince her to do surgical staging, so she underwent end to endobronchial ultrasound. Which in addition to the known cancer here, demonstrated a lymph node involved here in the hilum. Other mediastinal nodes were sampled and were clear. So at this point now, we had her as a stage two cancer.
Because of the demonstration of stage two cancer, she was referred back to our MMC and we were able to convince her that she should undergo surgery. She underwent a robotic left upper lobectomy, where we resected the entire upper lobe. We also harvested all these mediastinal nodes, including the AP window node which can't be biopsied from endobronchial ultrasound.
This node was also involved, so she ultimately proved to have a stage 3A non small cell lung cancer. You remember her PET scan staged her as a stage one. So we represent her tumor board, she tolerated her surgery well, and then underwent post-operative adjunct chemoradiotherapy.
There are a lot of studies that compared staging apparatus or modus. I'll just do one summary presentation. A publication in Chest in 2018 that basically showed that CT scan alone was about 60% accurate. PET scans and CT scans together, somewhere around 80% accurate.
You can get up to about 90% when you add endobronchial ultrasound. And then resection, of course, is the gold standard, or 100%. I think we've discussed now the benefits of surgery compared to SBRT, that surgical staging is often necessary to get a truly accurate stage.
I thought I'd talk further about what I consider to be a good cancer operation. The first is that the cancer has to survive surgery and recovery. The second affects, or concerns, how the surgery is done. Basically, there need to be complete surgical resection and signs that margins are negative, the nodes are all resected, and the patient should have a lobectomy, ideally, if their lung function allows.
All right, so how does minimally invasive surgery impact this definition? Well we know that minimally invasive surgery decreases perioperative risk, diminishes pain, and can probably broaden the application of surgery to frailer patients. However, the process of minimally invasive surgery, whether it's orthoscopic robotic, makes getting negative margins a bit more difficult, in some cases.
And so the good surgeon is choosing which approach, and is ready to change his approach in order to give a patient a good operation.
So I just want to briefly show two patients that were evaluated at other facilities and then got to us. This is a 77-year-old female with a significant smoking history, relatively borderline lung function with an FEV1 of 1.77 liters and diffusing capacity of 65%. She had these two lesions that were both biopsied and proven to be malignant.
She was evaluated by a non-Baptist surgeon in Miami and told that she shouldn't have surgery and no one else should. We evaluated her in her multimodality clinic, obtained quantitative perfusion studies as well as cardiopulmonary exercise testing, and got a PET scan which showed no activity elsewhere.
She underwent first endobronchial ultrasound, where all her notes are negative, and then underwent a thoracoscopic left upper lobectomy. The path showed two separate 3.5 and 3.8 centimeter cancers in the left upper lobe. They were both adenocarcinomas, nodes and margins were all clear.
She was in the hospital three days, and is alive at nine months with no evidence of disease. So I'd argue that in this particular case, the other surgeon didn't consider the benefits of minimally invasive surgery.
On the other side, though, it's possible to try to extend it too much. This is a 75-year-old male with a smoking history who was found to have a lung cancer. This was biopsied, it was an adenocarcinoma. You can just appreciate in the anterior mediastinum, he also had an anterior mediastinal mass that was biopsied and proved to be a thymic carcinoma.
We evaluated in multimodality conference, felt that the thymic cancer was large and that he should get neoadjuvant therapy prior to resection. However, he got a second opinion at another hospital, underwent a combined robotic thymectomy with robotic right upper lobectomy.
He had a completely resected lung cancer, but multiple positive margins in the thymus. He received an adjuvant radiotherapy, and then his PET scan at 12 months shows uptake in the left anterior mediastinum, as well as the hilum of the right lung. And he's undergone an endobronchial ultrasound, and these nodes are now involved with thymic carcinoma.
So a thorough operation in lung cancer is typically a lobectomy. And again, to go back to a crude drawing from the first patient, if this patient had had a wedge resection without appropriate staging, we would have missed that she had stage nodes in the hilar position, as well as nodes in the AP window, which made her a stage three cancer.
That's why a wedge resection is not a good operation, and it may also speak to why SBRT fails sometimes because there's so many cult stage twos and stage threes.
Multiple studies have shown that lobectomy gives a higher five-year survival over wedge resection, and we're-- remember, we're not talking just about control, but about pure. So in conclusions, radiologic staging, including PET and CT, is wrong in about 20% of patients.
The best surgery is lobectomy with mediastinal lymph node dissection. Lesser resection should be done in limited cases. A good surgeon chooses the appropriate approach, whether it's minimally invasive or open. The local control refers to the primary legion, not to the rest of the lung or distant recurrence.
All art series that have compared SBRT in surgery and found large differences that favor surgery. And finally, complete resections can be done with all minimally invasive techniques for many patients. The best surgeon selects appropriate cases for each approach.
I think that's it. Those are the references I have if you want to chase any of those down. Thank you very much.
DR. MARK DYLEWSKI: Thank you very much, Dr. Roberts. Excellent talk. Our next speaker is Dr. DeRosimo, he's a thoracic surgeon at the Miami Cancer Institute. And he will be covering the ABCs of the state of the art of minimally invasive robotic surgery for lung cancer. Welcome Dr. DeRosimo.
DR. JOHN DEROSIMO: Good evening. Thank you, Dr. Dylewski. Thank you to the continuing education program at Baptist Health for inviting me to speak this evening on the ABCs of robotic assisted pulmonary resection. I'd also like to Thank Dr. Dylewski for assisting with some of the slides and videos that I will be showing this evening.
These are my disclosures.
Tonight, I'll review with you the basics of performing a robotic assisted lung resection. I want to emphasize that a robotic resection is different from a VATS approach in that it is complete port access surgery. There is no utility thoracotomy through which the surgeon operates.
Everything is done on screen through port access. We will review the equipment used, and how it is set up in the operating theater. We will review positioning of the patient. We'll review the different approaches for robotic lung surgery. We'll go over the position of the ports for access to the lung and hilum.
We'll review the main steps in surgery as the surgeon conducts the operation. I'll then review the handling of a technical problem, specifically a vascular injury.
These are the components of the robotic system used at Baptist Health of South Florida. We use the Da Vinci robotic system from Intuitive Surgical. The robot has three components. The surgeon sits at the surgeon console, seen here on the left of the screen. From that console, the surgeon directs the instruments.
The operating instruments are loaded on the patient cart, seen on the right of the picture. This cart is placed over the patient and the arms are inserted into the body cavity for surgery. The vision cart in the center of the photo contains an assistant screen, the camera interface, electrosurgical control units for monopolar and bipolar cautery, harmonic scalpel, and vessel sealer devices.
These three components make up the integrated system.
The operating theater is set up in this way for robotic pulmonary surgery. The personnel are in the pink circles. Once the robot is docked to the patient, the surgeon takes a seat at the surgeon console. The surgical assistant and scrub tech are on opposite sides of the patient field.
Each one will have access to the robot arms for changing instruments. We positioned the robotic patient cart to the patient's left side and the vision tower at the foot of the bed.
A few notes about the anesthesia management. We use double lumen endotracheal tubes for lung isolation and single lung ventilation during surgery. The experience level and comfort level of the anesthesiologist will usually determine whether a central venous catheter, arterial line, and Foley catheter are used during the operation.
We've tried to get away from the routine use of arterial lines and Foley catheters in lung surgery, with varying degrees of success depending on which hospital and which anesthesiologist is doing the case. In high volume centers for thoracic surgery, routine use of arterial lines and Foley catheters has been eliminated.
The positioning of the patient is similar to standard thoracotomy. We use a lateral decubitus position. The patient is secured into place by a bean bag. The table is flexed to open the rib spaces. We also use an inflatable pressure bag under the waist. This is inflated prior to activating the bean bag, and it gives us a little bit more elevation of the patient's waist.
This provides easier access for the assistant port instruments, especially in patients with wider hips.
The surgeon has a choice of docking three robot arms or four robot arms for the operation. A three-arm technique is easier to learn since there is no switching between arms at the surgeon console. Also, there may be more freedom of movement with the arms since the ports are slightly further apart.
With the three-arm technique, the surgical assistant provides retraction of tissue. During the four-arm technique, the surgeon uses the fourth arm to set the retraction.
The surgeon starts the operation by placing the ports and docking the robot arms to the ports. The anterior port is placed first. This is also the port that will be used for stapling later in the operation. I usually place the anterior port at the sixth interspace anteriorly, for both right and left sided operations.
The only difference is for a right middle lobectomy, where I like it better in the fifth interspace, which seems to give me a better approach to stapling the middle lobe artery, vein, and bronchus. The other arms are then visually guided depending on the position of the major fissure.
The ideal placement is one interspace below the fissure, which usually puts the arms in the seventh interspace. The assistant port is in the anterior axillary line at the ninth or 10th interspace. The 10th interspace has the advantage of having soft tissue that can stretch when the specimen bag is removed.
But I have had a couple of post-operative hernias at the 10th interspace when I use that spot.
Since adjusting my position-- my assistant port to the ninth interspace, I have not had any post-operative hernias at that site. Again, we see initial access via the anterior port site. Insufflation with carbon dioxide and then placement of the remaining ports using visual identification of the anatomy of the major fissure.
In the forearm technique, the anterior port is usually a little lower at the eighth interspace, with the remaining three arms along the seventh interspace. The assistant port at the bottom is shown in the 10th interspace at the insertion of the diaphragm to the chest wall.
It is important to keep in mind the objectives of a complete robotic port access lobectomy for maximum patient benefit. The operation is performed with trocar access only, and no utility thoracotomy. The robotic operation is performed the same way as an open lobectomy, and follows strict oncologic principles of anatomic dissection.
There is individual dissection and ligation of the vascular and bronchial structures. The surgeon performs a complete lymph node dissection and the specimen is removed without thoracotomy or rib spreading.
A note about stapling. The surgeon can use either a handheld stapler or the robotic stapler. The handheld stapler would be positioned and fired by your assistant standing at the field, while the robotic stapler comes in on a robot arm. The robotic stapler is guided by the surgeon at the console.
The robotic stapler is more steady and stable compared to a handheld stapler controlled by your assistant. I, as the surgeon, have less stress when I'm in control of the stapling instrument and it isn't bouncing around on the field on an 18 inch handle held by my assistant through a port site.
The operation is performed in a standard fashion. The operative field is exposed. A complete lymph node dissection is performed. The fissure or fissures are divided. The surgeon performs dissection with ligation of the vascular and bronchial structures for the particular lobe being removed.
We're now going to look at some videos showing parts of a right upper lobectomy from start to finish. Here on this video, we see the start of the operation and the division of the inferior pulmonary ligament. The level nine lymph nodes at the inferior pulmonary ligament are harvested at this time.
After this, I usually divide the anterior plural reflection at the hilum, harvesting any hilar lymph nodes in the area.
Now to the posterior aspect of the hilum. The subcarinal space is opened, and the level seven subcarinal lymph nodes are dissected out of the subcarinal space. You will see the esophagus posteriorally, the right main bronchus anteriorly, and the pericardium covering the left atrium deep to the lymph node packet.
Sometimes you will find large bronchial arteries in this area. They can be clipped for control. The lymph node packet is removed intact. For bulky specimens, the lymph node packet can be placed into the cut finger of a glove for intact removal.
We now move to the paratracheal lymph nodes at stations 2R and 4R, the upper and lower right paratracheal lymph nodes. The dissection starts with division of the pleural reflection of the superior aspect of the hilum, separating the lower paratracheal lymph nodes at the tracheal bronchial angle from the underside of the azygos vein and from the right main pulmonary artery and distal trachea.
This dissection is carried superiorly along the trachea and superior vena cava to the upper paratracheal area. You will notice the small, rolled gauzes in the field. These are three inch Raytec gauze made into rolls and tied with silk ties. These gauze are very useful for holding retraction and for mopping up the blood in the dissection field.
Again, the specimen is removed intact.
Dissection is then carried along the posterior and lateral aspect of the bronchus intermedius. This will allow us to take the interlobar lymph node, level 11, and to set up the division of the common bridge of tissue in the posterior major fissure.
Then dissecting along the interlobar pulmonary artery in the fissure, we identify in that same plane to the posterior hilum on the bronchus intermedius. You'll see that we use a modified Foley catheter as a guide for the stapler going through the tissue. The stapler then ligates and divides the common tissue in the posterior major fissure.
The posterior ascending branch of the pulmonary artery to the right upper lobe is then dissected. It is ligated and divided by a vascular stapler.
The right upper lobe bronchus is dissected next. We get around the bronchus and then ligate and divide the bronchus using a thick tissue stapler.
We then dissect along the pulmonary artery, elevating the lobar lymph nodes onto the specimen. This dissection is continued to the truncus anterior branch of the pulmonary artery. This arterial branch is isolated and the plane developed between the truncus vessel and upper edge of the superior pulmonary vein.
The arterial branch is ligated and divided with a vascular stapler. The minor fissure is then divided using a thick tissue stapler.
A heavy specimen bag is then rolled tightly and brought into through-- brought in through the assistant port. The bag is unrolled inside the patient and the specimen is placed into the bag. The bag is pulled closed. We then might use a commercially available pulmonary sealant along the fissure and dissected portions of the lung parenchyma.
A chest tube is placed, the lung is re-expanded, and the robot is undocked from the patient. The assistant port site is enlarged slightly to accommodate the removal of the specimen in the bag. The specimen is removed and frozen section obtained on the bronchial margin.
The wounds are then closed in layers with absorbable suture.
I'll touch for a moment on how I handle a vascular injury, if it occurs. First, just hold pressure to control the bleeding. The rolled gauze is good for holding pressure. Or the lung tissue itself can be held over the injury to stop the bleeding. The pulmonary artery and veins are low pressure systems, so either of these maneuvers should provide control of the injury.
Suction brought in by your assistant will keep the field clean so that you can see. The injury can then be controlled with surgical clips, or repaired with a suture, depending on whether or not it's part of the specimen. I use a Pledgeted 4-0 Proline if it's an arterial injury in an area that is not part of the specimen.
If I need to open to control the injury, I'll place the anterior robot arm down, holding pressure, and remove the other robot arms so that I can perform a thoracotomy from behind the patient. Once I'm inside and I have a sponge stick ready to control the injury, I will remove the robot, undock, and then proceed.
Here's a video of a vascular injury.
The gauze is placed over the injury for control. Suction is brought in to keep the field clean. The injury is then controlled with surgical clips.
Here we have the closure and the incisions from a complete robotic port access lobectomy. Thank you very much.
DR. MARK DYLEWSKI: Thank you, Dr. DeRosimo. It was an excellent summary of a very complex technique. The next speaker is myself. I'll just let the video play, and it'll introduce me. And then we'll move on. Thank you very much.
Good evening. I'd like to thank the planning committee for the opportunity to speak to you all tonight at the MCI Baptist Health lung symposium. Tonight I'll be talking about robotic assisted pulmonary resection management of locally advanced and complex pulmonary malignancies using a surgical robotic technique.
I do have a few disclosures. I do serve as a clinical educator for a number of companies listed here.
Today, the treatment of lung cancer has been changing quite rapidly. And you can see a sort of a short timeline of how we've moved from an open approach to treating lung cancer to a more minimally invasive approach. And now we've moved into the digital age of what I call surgery.
Competing with technologies such as Cyberknife and transthoracic ablative technology and the advent of the robot and the adjunctive technologies that can be utilized along with the robot has really improved our abilities to manage patients using a minimally invasive technique.
And dealing with a very complex surgical disease that perhaps we were not able to manage using a minimally invasive technique. And I'd like to talk about some of those advantages and the reasons be-- the reasons why I believe there are advantages over the standard open technique and the traditional, what we call, minimally invasive technique.
Currently, the adoption of robotics is developing quite nicely. Today we are seeing about 25% of all lobectomies performed in the United States being performed robotically. This compares quite favorably to the open approach and the VATS approach.
The VATS approaches remain relatively stable, around 35% to 40% over the last decade or more. And what we're seeing now is more and more surgeons who used to do open surgery are adopting robotics because it tends to be a lot easier to integrate into your practice than traditional VATS or laparoscopic surgery.
So there's certainly an advantage to this patient population as more and more patients are being offered the minimally invasive approach. Whether that's done with traditional VATS or robotics. Now VATS, in my opinion, is a technology that has a limitation-- a significant limitation.
And [INAUDIBLE] in 1997 was-- published a paper looking at the adoption rates of traditional minimally invasive surgery for thoracic surgeons, both in the academic and the private practice setting. And it was a survey of over 300 practicing high volume surgeons as to-- the questions as to why VATS was not adopted in the majority of patients that they were operating on.
And the issues were multifactorial. Oncological control certainly was an outstanding question as to whether VATS was equivalent to open, limited instrumentation. Certainly there's been very little advancements in traditional VATS instrumentation for the past decade or so, making this surgery pretty much a counterintuitive type-- a very difficult, challenging technique to integrate into a practice without spending an exorbitant amount of time learning the technique.
Operative times tended to be quite long. And certainly, there's insufficient training once you've left your respective training program if you're out practicing as an independent surgeon. It's very difficult to train yourself in this technology. Later on, Dr. Demmy in 2008 recognized, in a retrospective analysis of online data, that traditional minimally invasive lobectomy demonstrated equivalence improving long term survival in early stage disease.
But he indicated in his conclusion that the technical inadequacies of minimally invasive lobectomy should be amplified for more advanced cancers. And I would like to talk in detail about that open question, as to whether traditional VATS is an oncological equivalent operation compared to open.
And then we'll compare it to what we are seeing with the newer technology robotics.
If you look at Dr. Boffa's study, published in the Annals in 2012, he compared STS database of open surgeries to VATS surgeries. And you can see over time, videoscopic surgeons tended to be able to develop a better technique of nodal dissection compared to open, as their experience improved.
And the conclusion of the paper was quite interesting. He said during lobectomy or segmentectomy for clinical and zero lung cancer, mediastinal lymph node evaluation by VATS and thoracotomy results were equivalent in upstaging. In contrast, lower rates of N1 upstaging in the VATS group may be indicative of variability in the completeness of the peribronchial and hilar lymph node evaluation.
And this makes a lot of sense because it's much easier to use chopstick type instruments to dissect around the main bronchus or the large pulmonary arteries as compared to the central hilar structures, the pulmonary artery, because that's where the N1 lymph nodes lie.
And so using sort of traditional videoscopic surgery, it requires a higher level of skill and a more-- certainly a much more complex nerve-wracking procedure to dissect off N1 lymph nodes around the pulmonary arteries in the hilum as opposed to the N2 lymph nodes that are usually around the bronchus or the trachea.
So there may be a reason why we're seeing a lower rate of upstaging in N1 lymph nodes when VATS is used in this patient population.
If you look at another study by Merritt, he also compared VATS lobectomy to open lobectomy, and he showed that there is a significant percentage of upstaging from N0 to N2 or N2. Almost 25% in the open group versus the VATS group. So if you choose to use VATS, there's a potential that you'll be under staging 15% of your patient population by doing the surgery with traditional VATS as opposed to open.
And thus, those patients would not benefit from adjunctive therapy such as chemotherapy and radiation. So you really need to be cautious, in my opinion, in utilizing VATS for those patients that are truly clinical N0. Those patients that have a higher risk of having an occult metastases in the lymph nodes or more advanced cancers.
One should really ask whether traditional VATS is an appropriate operation for those patients who have more advanced disease or harboring a higher risk for occult metastases. And that's where robotics may play a significant role.
Now I'm going to talk a little bit about how the complexities of treating locally advanced disease. This is a challenging group of patients. It involves a heterogeneous population of patients with hilar and mediastinal involvement, central locating tumors. They may be very proximal to pulmonary vessels and the proximal bronchus and trachea.
And these patients often require induction therapy, which increases the risk of fibrosis and desmoplastic changes around these structures, making it sometimes very challenging to do these cases through a minimally invasive approach.
Now there has been some publications in the past, mainly out of the D'Amico group at Duke, and this is a small series of patients-- 97 consecutive patients with non-small cell carcinoma-- that received induction therapy and were approached videoscopically. 12 patients were successfully resected using traditional VATS approach.
67 of those patients received induction radiation therapy. And only one patient was converted. So in their conclusion of their paper, they found that utilizing minimally invasive surgery for patients who received induction therapy, who had locally advanced disease, was safe and had certain benefits with reduced length of stay of the hospital, overall complications in the patient population.
So they recognized that minimally invasive surgery was advantageous in this patient population. But it does not answer the question as to whether it's an equivalent oncological surgical procedure in patients with advanced disease compared to an open traditional approach.
Now if you look at the data using robotic lobectomy-- and I'll talk about some of the largest trials that have been run in the recent past few years. This is a trial that was recently published from the PORTaL Investigation Group. It was an analysis of over 6,300 cases from 21 high volume institutions.
And it was presented at the STS annual meeting in 2020. And you'll find when you compare traditional VATS to robotic lobectomy, there is a much higher conversion rate in the traditional VATS approach than the robotic approach, which may suggest that the traditional VATS approach may have challenges-- may not be able to perform to the level of resectability compared to an open approach or robotic approach.
And you can see here in this illustration that the stage two patients, which are the patients that have typically N1 lymph node involvement, may be a challenge addressing them with traditional VATS approach. Whereas robotic had a better-- lower conversion rate. And also the stage 3A patients had a much higher conversion rate, and those patients typically have more desmoplastic changes and fibrosis and invasion into more central structures.
Again, suggesting that the VATS approach may be inferior to robotics or open.
Now we also would like to spend some time talking about one of the largest long-term survival series, published by myself and Dr. [INAUDIBLE] in 2018. It incorporated almost 1,400 consecutive patients. And you can see that the outcomes were quite good. Operative times were quite respectable at 136 minutes.
There was a 9% conversion rate for tumor-related reasons. Very small conversion rate for bleeding, about 2%. And the length of stay and the major morbidity and mortality was quite acceptable and comparable to either open approaches or VATS approaches. But what we were surprised to see was that the five-year stage specific survival for each stage tended to compare favorably to either anything previously published for the VATS patients or for open patients.
So this study certainly deserves more investigations and follow up, but it would suggest that perhaps robotics allows one to do a more thorough lymph node dissection, a thorough R0 resection, and may influence long-term survival.
So the study basically showed that the outstanding stage specific survival results are at least comparable, and maybe favorable. At this time, follow up data is superior to most other previous reported studies either using open or traditional VATS approach.
Our own data from our institution looked at 1250 cases produced between January 2007 and June of 2018. And in that category of patients, we collected about 214 patients with advanced stage disease. The outcomes in this group are quite favorable. All of this-- these patients received typically upfront adjuvant chemotherapy and radiation.
And the conversion rate in this patient population is quite respectable, similar to what we saw in the larger publication by Dr. [INAUDIBLE] at about 2.5%. And very few patients were converted for bleeding. They were mainly converted for two-- because of tumor reasons.
Either it was a very centrally located tumor and we had difficulty completing a sleeve lobectomy, or we were trying to spare a lobe that eventually needed a pneumonectomy. So this technique has been proven to be reliable, safe, and reproducible. You can see here in this 214 case series, there were 65 patients with stage 3A disease that received full dose radiation pre-operatively.
The median tumor range from 0.7 centimeters to 11 centimeters, so quite a challenging group of patients to treat. And what was surprising is that our overall morbidity and mortality was quite acceptable, with the overall morbidity being 26%. The major morbidity being 5%.
And you can see here in this illustration that the common morbidities included an air leak that lasted more than six days and an effusion requiring a thoracentesis typically after surgery. Arrhythmias, about 3%, and pneumonia rate about 5%. So these complications in this very challenging group of patients you can see is quite respectable.
If you recall how these patients responded to a previously open technique to treat locally advanced disease, these patients often had multiple complications postoperatively, spent a long time in the hospital recovering. Often repeat procedures such as bronchoscopies, readmissions to the hospital or to the ICU.
So robotics and minimally invasive surgery in general for this patient population has been a positive attribute, in my opinion.
This is just an example of a patient who had a very large, poorly differentiated neuroendocrine tumor in the left upper lobe. We treated her with preoperative chemotherapy and radiation. And you can see here that we're approaching her robotically. It's a very centrally located tumor.
We're certainly going to expect some significant desmoplastic changes around the central hilum. And you can see here as we're dissecting around the bronchus of the left upper lobe, we're unable to free up that large lymph node that was predominantly in the super hilar location.
And this just demonstrates the flexibility of robotics being able to perform very complex maneuvers, such as dividing the [INAUDIBLE] to be able to get better access to the main structures, namely the super hilar pulmonary artery structures, to safely divide them without getting into too much difficulty.
And without the availability of robotics, using traditional [INAUDIBLE] surgery, this type of surgery would be very difficult and challenging to do for most thoracic surgeons, even those of us that had experience with traditional VATS.
So in summary, conventional total endoscopic video-assisted anatomical resection remains technically demanding. I think advancements in robotic assisted platforms have made it possible and reliable to perform a complete port-based pulmonary resection, even for more advanced disease, not only just early stage disease.
Robotic assisted and topical lung resection is feasible and safe. It's been proven to be favorable to historical series published on conventional open technique or VATS technique. Robotic assisted lobectomy is associated with low morbidity, low mortality, and a short length of stay.
And robotic assisted pulmonary suction has a wider clinical utility. Those patients who have larger masses, more advanced disease, or lymph node involvement can be managed using a robotic minimally invasive technique. And one-- if one chooses to adopt this technology into their practice, if you're an open surgeon, certainly you're going to be able to manage a wider population of patients, those with more advanced disease, with a minimally invasive technique.
And I find that managing patients minimally invasive as much as you possibly can will improve the outcome, reduce the complications. And it has also been demonstrated that the utilization of minimally invasive surgery reduces the overall cost to the health care system.
So there's certainly an advantage to try to introduce minimally invasive techniques to as many patients with locally advanced disease or early stage disease for our lung cancer patients.
Thank you very much for the opportunity to speak this evening. Our next speaker is Dr. Rupesh Kotecha. He's the chief of radiosurgery in the field of thoracic oncology, and he'll be speaking about the treatment of early stage and advanced lung cancers, the role of ablative technology. Thank you, Dr. Kotecha.
DR. RUPESH KOTECHA: I want to thank the organizers for having me here today. My name is Rupesh Kotecha, and I'm a radiation oncologist in the Department of Radiation Oncology at MCI. Today I will be speaking on treatment of early stage and advanced lung cancer: the role of ablative technology.
These are both my personal disclosures, as well as my institutional research funding. As well as from a conference perspective, I'm going to discuss some of the equipment or brand names in our department, but this is all an educational effort to explain how patients are triaged across the different technologies at our institution.
These highlight examples that we have made for patients that we have treated in the clinic, but do not represent the only acceptable option for any particular patient.
So this is our beautiful Cancer Center here in Miami. And what's really remarkable and unique is that on this first floor, we essentially have one of every single radiation machine there is, so we can personalize the radiation therapy treatment options for every patient that we see in the clinic.
Really, all of this technology is here to employ the optimal modality for any patient, given any diagnosis. Today, we will actually discuss in detail how we use these advanced technologies for patients with early stage and locally advanced lung cancer. Now under the hood of each of these machines, just like under the car hood, there are differences in what are called the inter-fraction, intra-fraction.
Those are imaging that is occurring before and during each radiation treatment. There's differences in the radiation type that each machine has. And this basically leads to ideal clinical indications for each of the technologies that we have at our center. This is actually complicated enough that we have an established workflow in place, in which a patient is seen in consultation.
We would create a directive as to how we want to simulate and treat a patient. This undergoes a multidisciplinary prospective peer review, which is actually very unique to our institution. The only one in, really the entire world, that does this. Then the patient undergoes simulation.
We delineate the target volumes and the organs that are at risk close to that target. We also review that contour set. We then do treatment planning. And then that is approved by an individual physician and then peer reviewed with another physician team. This undergoes quality assurance, typically by our physicist.
And then the patient actually delivers this radiation therapy treatment. This is an established workflow that we have, and our processes have been published in the peer reviewed literature.
Now for a patient with early stage lung cancer being evaluated at our center, I can think about five different radiation therapy machines that we could potentially treat that patient with. And as we are seeing the patient in clinic, as we're gathering the information from the shared medical record for that patient, as we are seeing them and doing a physical examination or looking at the imaging studies, we're processing all of this information for each patient.
They then be able to triage them appropriately to the machine of choice, which would be best to treat their cancer. In some circumstances, there is more than one machine that could be potentially used for each individual patient. If this circumstance or scenario arises, then we can do what's called a comparative treatment plan, and we can generate different treatment plans across the different platforms and do comparisons between them.
And essentially, at the end of the day, we want to treat the patient with the best quality plan.
Now for the purposes of the discussion today, for early stage lung cancer, I'm going to review one of the newest machines that we've added to our portfolio, which is an MR linear accelerator. This was the second machine in the country, the third in the world to be installed at any facility.
Essentially, the workflow for this, or as a patient comes into the clinic, they undergo an MRI scan in our department. That's very unique. Then they undergo a CT scan. That's what we use to calculate radiation dosimetry. We also do what's called a four-dimensional CAT scan.
That means that we watch the tumor as a patient breathes. The physician then does the target volume delineation. We want to delineate that target, as well as the nearby organs at risk. That undergoes planning and QA. And then when the patient's actually receiving their treatment, they undergo an MRI at the time of treatment in which we can visualize that tumor. And then the patient is actually treated with radiation.
Again, this is all non-invasive. The patient does not see anything, they do not hear anything, the machine does not touch them at any point. In fact, this allows us to see a tumor, track a tumor, and treat a tumor. And we can actually do this in just even one single fraction or a single treatment.
And we triage patients specifically to this platform, for this particular purpose. We can visualize the entire treatment and the patient themselves can actually visualize this as well.
This is a study that we published in the peer reviewed literature showing our single fraction radiation therapy workflow for patients with early stage non-small cell lung cancer and how we do this safely and effectively.
Now there is an established workflow for how this entire process arises. First, at the time of the treatment, I need to identify a target for anatomical tracking. That could be a region of interest to be treated, which is typically what we use for lung cancer patients, but could also be a critical structure that one wants to avoid treating.
For example, the spinal cord. We then create a boundary to identify a tracking region, and then we are able to visualize the tracking algorithm as it deforms the anatomical target and each subsequent frame. In the next slides, I'll actually show you visually of what this looks like.
Then treatment occurs if the target is within the boundary. On the other hand, radiation is paused if the target moves outside that boundary.
So let's walk through a case example here. This is actually one of the first patients that we treated on the machine. A patient who had a large metastasis to the lung from a previously diagnosed colorectal cancer. As you can see here, this is a large, left, lower lung tumor.
As you can see here on this sagittal slice, all of the radiation lines are essentially centered along that area and therefore we're sparing the adjacent organs such as the heart, the esophagus, the spinal cord, and the right lung. The prescription dose for this patient is 60 gray.
Now at the time of each treatment, the tracking structure is defined, as you can see here. And then there's a boundary that is placed outside of that. And essentially, this can be adjusted each day by the physician and you can evaluate the tracking ability. Essentially, the treatment occurs if the tracking structure-- the tumor in blue-- remains inside the boundary, which should be the radiation isotopes line that we're using to treat.
This is an example of an actual treatment.
In this video, you can see that the patient is breathing in and out. You can see the heart beating and the diaphragm is moving. If that tumor is moving outside of the red, the target outside of the boundary, then the radiation would be paused. So essentially, this is the beginning of the treatment before any radiation would actually be delivered.
The patient is breathing in and out and getting ready to then hold their breath for their actual treatment. This allows the patient to have the least amount of lung exposed to radiation or irradiated. So now you can see the patient is in breath hold. Their diaphragm actually remains stable, their heart is beating.
And during this period of time, that target, that blue tumor, is within the boundary, that red circle. So therefore, the radiation treatment would be occurring.
We use this for patients who have very high risk tumors in critical locations. So this patient essentially has three different reasons why they would be considered high risk. It's a 65-year-old with a very large tumor, as you can see visualized in this right upper lung. Number one is tumor size.
Number two is proximity to central organs at risk. This tumor is very close to, and really touches, the edge of the trachea. It's also very close to the esophagus. So those are ultra central structures. The third is that the patient has severe COPD, as you can see on these CT scan both axially and coronally.
This patient could not have surgery given the high medical and operability, or risk for surgery. This patient was treated to a dose of 40 gray. Given how close it was to the central structures, this was all done in five treatments, or five fractions, of radiation therapy. These are the scans before radiation therapy, as I showed before.
And then on the right, the post-radiotherapy or post-treatment scans. As you can see, showing an excellent response to treatment. On the corresponding axial slices as well, you can see a very large tumor. It essentially melted away with the radiation therapy treatment.
Now stereotactic radiation therapy, as I mentioned, is non-invasive treatment. And although it is primarily used for patients who are medically inoperable or medically high risk, there is consideration about what the long-term outcomes are of this treatment, as well as application to patients who are considered moderately high risk for surgery.
This is the most recent data that was published in Lancet Oncology from the M.D. Anderson group, in which they looked at patients who received stereotactic radiation therapy-- these are in red-- compared to a cohort of patients that were treated with surgery at the same institution.
So based on this propensity match score analysis, you can see here that the three year overall survival was 91% in patients who underwent stereotactic radiation, as well as those patients who underwent surgery. Similarly, at five years, you can see it's 84 versus 87%.
For the cancer specific survival, you can also see that they are very similar at three and five years in comparison of stereotactic radiation therapy, again in red, and surgery in blue.
One of the unique features about using this machine is that there's an adaptive process that also can be utilized. What if the anatomy changes from when we did all of the radiation therapy treatment planning, which could be a week or week and a half ago, until on the day of actual treatment?
So as we reviewed before, the workflow would end with the MRI use for adaptive planning, and then the patient would actually be treated. In certain circumstances, we can actually re-contour on the MRI scan the new tumor, the new target, any changes in the anatomy. Our physics team can actually adapt the plan and do quality assurance on a new radiation plan adapted for the anatomy of the day.
We would then do a [INAUDIBLE] MRI scan to visualize the tumor, and then the radiation therapy is then delivered. So let's see what this could look like.
So this is a simulation CT scan from a paper from the WashU group. You can see the tumor in the left upper lung in blue, and then the esophagus is marked by the E, which is in green.
If you look at when the patient's actually being treated with radiation therapy, the esophagus has shifted over to the left. It's in closer proximity to the tumor. So if you use the original radiation plan for this patient, you would think that you're giving a lower dose to the esophagus.
But in fact, when the patient's actually getting treated, they're getting a higher dose than what was anticipated. Look at the second fraction for that patient. Now the esophagus has shifted over, and is essentially touching the edge of that tumor. At this point, you're now essentially giving the esophagus much more radiation dose than originally anticipated.
What's unique is that on this machine, you can actually do a brand new radiation therapy treatment plan, where you can adapt the treatment knowing that the esophagus has changed positions. So therefore, you can actually bring the esophageal dose down. So this is where the esophagus dose would be if you treated the patient with the old radiation therapy plan with the new anatomy.
But if you actually replan the patient, you can bring the esophagus dose down from, say 47-ish, down to about 35-ish. This is a substantial reduction dose to the esophagus and would then correlate with a substantial reduction in toxicity associated with the treatment.
At the same time, you can actually boost the dose to the tumor based on the new anatomy. So it's not just a dose reduction, but potentially dose escalation. So for example, if the maximum dose to the tumor previously was below 60 gray, you can now then push the tumor to receive more than 60 gray by using or taking advantage of the current anatomy.
We are about to study this prospectively in a multi-institutional study called the lung star trial. This is a clinical trial for patients who are medically inoperable or medically high risk who have lesions that are deemed ultracentral or central, as defined according to their proximity to a lobar bronchus, the main stem bronchus, trachea, or esophagus.
These patients will be treated with stereotactic radiation therapy on the MR linear accelerator with adaptive treatment planning based on the anatomy of the day.
And then finally, I wanted to touch on another technology which is unique. Again, the use of proton therapy for lung cancer. Now there are many rationales to consider proton therapy for patients who have lung cancer. Could be to reduce the dose to normal tissues. It can safely allow for treatment of tumors that are close to critical organs that could potentially not be treated with photon therapy, which is all the traditional forms of radiation therapy.
It can also more safely allow for dose escalation and more safely allow for retreatment of locally recurrent tumors that could previously not be treated with photon therapy. There are a number of potential benefits, including reduced treatment toxicities. You could offer a patient a chance of cure not otherwise achievable with photon therapy.
You could potentially improve tumor control, or even progression free survival. And therefore, this leads to a promise for the use of proton therapy.
Now for patients who have locally advanced lung cancer who are not going to surgery, our recent randomized controlled trials and now long term follow up from these studies have shown that the addition of immunotherapy to concurrent chemoradiation therapy have improved the overall survival of our patients, as you can see here on the blue curve compared to the placebo curve, which is patients who receive chemoradiation alone in orange.
And this improvement in survival has really been dramatic for our locally advanced patients.
Currently we're putting these two principles together in the non-operative setting for patients who have locally advanced lung cancer, and studying the role of proton therapy. This is the RTOG 1308 clinical trial, in which patients with stage two and stage three non-small cell lung cancer, again non-operable patients.
Typically these patients do receive adjuvant immunotherapy as per the standard of care, as I just mentioned. Their patients are treated to dose escalated radiation therapy. So instead of that traditional dose of 60 gray, we're now going to 70 gray. And these patients are randomized to receive either proton therapy or photon therapy.
This study is looking at the overall survival between these two patient populations, and a number of secondary endpoints including treatment related toxicity.
Now not all patients are candidates for concurrent chemotherapy. And for these medically high risk patients who cannot have surgery, who cannot have concurrent chemotherapy, we're also doing research to improve their outcomes. This is not something that is commonly evaluated, but this is commonly seen, especially in elderly patients.
So this is a clinical study that we are partnering with Memorial Sloan-Kettering for patients who have stage two, stage three non-small cell lung cancer. And these patients receive definitive thoracic radiation therapy and immunotherapy at the same time, and then receive adjuvant immunotherapy.
The primary objective of this study is to demonstrate an improvement in the two year progression free survival rate compared to our historical outcomes. So hopefully today I've given you a glimpse into two areas of advanced radiation therapy, both in the use of MR guided radiation therapy for patients with early stage lung cancer, as well as an introduction to the use of proton therapy, which is undergoing extensive research at our facility and across the country.
Thanks so much for your attention.
DR. MARK DYLEWSKI: Thank you very much, Dr. Kotecha. Very excellent, complex subject to cover in 15 minutes. Our next speaker is Dr. Andres Sosa. He's a board certified pulmonary critical care and fellowship trained interventional pulmonologist. He will be giving us a talk on the evaluation of pulmonary nodules and the advancements in diagnostic bronchoscopy. Welcome, Dr. Sosa.
DR. ANDRES SOSA: Good afternoon, everyone. In the next 15 minutes, I will be talking about the evaluation of lung nodules and explaining some of the advanced bronchoscopic techniques that we have to approach these lung nodules. I'll start by saying that I have no conflicts of interest to disclose regarding this presentation.
We will be talking about what the definition of the solitary lung nodule is, how do we approach them. And more specifically, we'll be going into navigation bronchoscopy. We'll be talking about the background of navigation bronchoscopy, why is it needed, what is it. And then we'll go specifically into electromagnetic navigation and the robotic platforms as well.
So what is a solitary lung nodule? It's an approximately rounded density within the lung, and it's usually less than three centimeters in size. It is completely surrounded by lung parenchyma, and it is not associated to any other abnormalities, such as athletic disease or pleural effusions.
When we approach a solitary lung nodule, we need to be aware of the characteristics that may portend a higher probability of malignancy. Some of these characteristics would be size. So the bigger the nodule, the more likely that the nodule is malignant. Morphology. So irregularly shaped nodules.
Spiculated [INAUDIBLE] with spiculated borders will also be more suspicious. The type of opacity, whether it's a solid, sub-solid, or a ground glass nodule may also have different implications.
There are well validated calculators that we can find online to do, let's say, a pretest clinical probability of whether a nodule is malignant or not. One of those tools you can see right now. Very simple to just click on a tool like this, and you can just put in different variables that refer to your patient and get a number back on what the approximate probability of the nodule being malignant is.
When we talk about the photo op of a solitary long nodule, we-- there are different guidelines that we can follow. One of the most widely used guidelines is the slicer society guidelines. It takes into consideration both the size of the nodule and whether the patient is high risk or not.
A high risk patient being, generally, somebody who's been a long time smoker or exposed to asbestos or a prior history of cancer, for example. Nodules that are eight millimeters, or bigger than eight millimeters, are the nodules we really want to look into more closely with either a PET CT or tissue diagnosis, or sometimes even going straight to surgery.
So based on what-- on the particular risk of our nodule, we are going to decide one of three things. Do we do a serial CT? Well typically to do a serial CT, we want nodules to be less than eight millimeters, so we're very low risk. When the nodules are anything more than very low risk, meaning low to moderate risk, that's where either a diagnostic procedure or a PET would come in.
We could also think about doing a diagnostic procedure when we actually need a diagnosis so we can start some form of therapy. Also when the clinical picture and imaging are not congruent with each other. Let's say a patient who's a very low risk patient who has a nodule that may or may not be malignant.
We want to sample it before we actually commit that person to surgery. Patient preference also very important. Some people are just too anxious to follow a nodule with CT and they want to get them sampled or they want to go straight to surgery. And the patient preference is always something we need to consider and factor in.
Whether the patient is a high surgical risk. Every time somebody a high surgical risk, we should think about perhaps doing a diagnostic procedure first. And then who goes straight to surgery? It's going to be whoever has a high risk nodule, meaning more than 65% if we're using one of these validated tools.
Or if the physician, who is accustomed to following lung nodules, deems a nodule to be high risk. This recommendations are based on the American College of Chest Physicians from 2013. They have not been updated. These are still the guidelines that we're going by.
Again, this is more of the same. So we have a patient who comes in with a nodule anywhere from eight millimeters to 30 millimeters. We assess the surgical risk. High surgical risk, well, if they're moderate or high risk, we think about doing a diagnostic procedure first. If they're very low risk, or maybe up to low to moderate risk, we think about doing CT surveillance.
However, if the patient is not a high risk to surgery, then we say, OK, very low risk. We go to a CT surveillance. Low to moderate risk, we may do a PET. Depending on the PET, we may decide on either doing a surgical biopsy, a non-surgical biopsy, or proceeding with surgery.
And for high risk nodules, we want to do a standard evaluation with PET and go to surgery, unless they're high risk for surgery. Then we would again be looking at a non-surgical biopsy first.
Now into navigation-- going into navigation bronchoscopy. So why-- what is navigation bronchoscopy? Navigation bronchoscopy means doing a bronchoscopy to get to the nodule with the help of some technology that's going to guide me to get there. And why do I need that?
Well if we did just conventional bronchoscopy, the sensitivity and negative predictive values are not nearly good enough. So conventional bronchoscopy is just not going to cut it. We might as well not do anything. How about fluoroscopy during bronchoscopy? Well, fluoroscopy is certainly helpful if we can see the nodule-- if it's large enough to be seen on fluoroscopy and if there's a direct airway straight into that nodule.
If we have to do many twists and turns, if we are beyond the fifth, sixth generation airway, it's very unlikely that we're going to be able to get there without some sort of guidance. And that's where navigation bronchoscopy comes in as well. How about if we just do a trans-thoracic approach?
Well, CT guided biopsies are very sensitive. The problem with CT guided biopsies is that they also have a high risk of pneumothorax. Up to 15%, 20% depending on which reviews you look at. Up to half of these patients will need a chest tube placed. And this is something to consider when we're talking about a patient population that already has a very low, very limited pulmonary reserve.
When we go endoscopically, as we talked about, we need some sort of guidance. We need to know how to get to the nodules, especially when we're talking nodules that are in the periphery, beyond the fifth, sixth generation bronchi. So here is where the navigation techniques come in.
We have virtual bronchoscopy, electromagnetic navigation bronchoscopy, and more recently the robotic platforms. When it comes to virtual bronchoscopy, what is it? Well, these are technologies in which we can use the patient's CT, grab, basically, the CD with the DICOM images, put it into the computer software.
The software will show us a map that we will accept or not. Basically we will choose a target, and then the software will generate a path towards that target that we will use during the time we're doing our bronchoscopy. Having said that, please note this is not real time.
This is another example of the virtual bronchoscopy technology. You can see there's a path made towards the target, which is the nodule. And in this particular software, the-- there's different-- there's a bunch of different pictures that we have to match with the image that we're seeing endoscopically.
So again, it's not real time, meaning we have a pre-formed path and we try to match it as we're doing-- as we're doing the bronchoscopy. Electromagnetic navigation gives us a little bit better guidance in the sense that it is real time. And please know that when I'm saying better guidance, I don't mean one technology's better than the other because there are no head to head studies to compare one technology versus the other.
Electromagnetic navigation is probably the one that's more widely used nowadays, and it is like a GPS system to get us to that nodule. And just like a GPS system, we have a destination and we have real-time feedback on where we are and how to get where we want to go.
So this is pretty much the flow of what the procedure would look like. First, we would put our CT DICOM images into the software. We will choose a target, and then we will go ahead and do our procedure. This is what the setup would look like when we're in the OR, or in the endoscopy suite or the procedure area.
It is good to note that we can also use fluoroscopy, and we can also use radial endobronchial ultrasound with these technologies. So this would be an example of a case. We have a left upper lobe lung nodule, and I'm going to use electromagnetic navigation to get there.
So I plan my procedure. And then during the procedure, I get feedback from-- real-time feedback that tells me where I am, and it tells me when I get to that nodule. Then after I'm there, how do I know that I'm really there? Should I just trust the software? Well, you could.
Or you could also do radial endobronchial ultrasound. So this is just an endobronchial ultrasound that I will advance through my working channel, and it's going to show me the actual nodule. So not only have I just navigated there, now I know for sure that I'm there because I can see it with the ultrasound. And then I can biopsy it and I can use [INAUDIBLE] to make sure I'm not moving.
This is just another example of one of the electromagnetic navigation platforms that we have. And then how about the yield? How good are these technologies? Because we saw that trans-thoracic approach, the yield is around 90% or sometimes even higher. How about electromagnetic navigation?
Well historically, and this is now for the past 15 years, every study that's been done has been shown approximately the same yield. Anywhere from 69 to the mid 60s to the low 70s. And the widely-- the biggest meta analysis done to date, very widely-- very widely supported and cited includes 39 studies with over 3,000 lesions.
And again, we see electromagnetic navigation bronchoscopy having about a 67% yield.
The most up to date study, and the most comprehensive study that's been done so far on electromagnetic navigation-- electromagnetic navigation is the Navigate study. Here, the yield was 72.9%.
So when we do this navigation bronchoscopy techniques, there are things that we need to consider when it comes to our yield and when it comes to planning for our procedure. So yields will sometimes vary depending on whether it's upper or middle lobe locations of the nodule, what the size of the nodule is, whether there's a bronchus sign within the nodule, whether we're using endobronchial ultrasound or not at the same time.
So different reviews yield different results.
The Navigate study, again the most comprehensive study that's been done, over 1,000 patients over a year, a full year follow up, showed us that the yield may actually be impacted in a positive way if we use less tools, so less than three tools. If lymph nodes are biopsied at the same time, if there's a bronchus sign present.
And this correlates with other studies also showing that the bronchus sign may be helpful, in terms of diagnostic yield. Whether multiple lesions were biopsied, and the total procedure time. Shorter procedure times usually portend a better yield.
It is a very safe technology. The pneumothorax rate is about 2.9% in the Navigate trial. And these are 2.9% pneumothoraxes that actually needed a hospital admission for observation or a chest replacement. And bronchopulmonary hemorrhage just need an inpatient observation, about 1.5%.
Respiratory failure, exceedingly rare, less than 1%. Now moving on to the robotic platforms. So the robotic platforms are basically the newest technology. The yield with electromagnetic navigation, as we talked about, is usually around 70%. However, some trials-- or some reviews, I should say, have shown the deal to be as low as 38.5%.
This comes from the Acquire database, which is a database of over 15 different hospitals of whatever-- of what's happening in the community, what is happening outside of clinical trials. So one has to wonder whether, when we're outside of the clinical trials, our yield is actually lower than that 70%.
So robotic platforms may be helpful in this regard. And we have two ways of-- two types of robotic platforms right now. We have one that uses electromagnetic navigation for guidance, and we have one that uses shape sensing technology for guidance. They both have the advantage of really having more steering capacities.
We can pretty much get into any area of the periphery of the lung, and we can orient our catheters pretty much into any-- into any direction. And we also have the capability of remaining that-- of keeping that catheter in place, of not moving the catheter. That's where that's in different instruments.
This is an example of one of the robotic platforms. As you can see, you have something that looks just like an Xbox controller, and you use this to orient your rope to guide your catheter towards the target lesion. And you can also use the guide-- the catheter that gets you there to advance a radial endobronchial ultrasound, look at the lesion, and sample it at the same time.
And in this particular platform, again, the patient is in an electromagnetic navigation field. And that's how we get real-time feedback from the software.
We have a retrospective post-marketing review of the electromagnetic navigation platform showing a yield of 69.1 to 77%, and this is-- we're talking about 165 patients so it's a fair number with a complication rate that's similar to conventional electromagnetic navigation.
And here, lesion size, density, lobar location or centrality actually did not affect the yield, which also speaks in favor of the technology being able to get in less accessible areas.
There's the BENEFIT trial, which is a larger prospective multicenter pilot and feasibility study, including 55 patients from five centers. The median lesion size was about 23 millimeters. And again, the yield was about 74.2%. So this is the median lesion size, it's very approximate to what we're going for in the-- with the conventional electromagnetic navigation technology.
And again, the complication rate, 3.7% pneumothorax comparable to other technologies.
Besides from electromagnetic navigation, there's also something called shape sensing technology. Here, the catheter has many, many small different sensors that actually can account-- can have real-time feedback in terms of the length of the catheter, how much of the catheter is actually in the patient, and where is the catheter, what's the orientation of the catheter.
And again, we are driving from outside and driving towards the lesion, getting real-time feedback. So the shape sensing technology, we have data from a single center in which data was collected prospectively and then analyzed retrospectively. These are 159 lesions, medium size, about 1.8 centimeters.
And we see that the diagnostic yield was about 81.7%. The lesion size and central location were associated with a higher yield. And again, the complication rate was quite low. Pneumothorax, about 1.5%. Overall complications, 3%. What's also exciting about navigation bronchoscopy is its future applications in therapeutics.
We can imagine actually not only localizing and diagnosing a lesion, but also treating it at the same time. And trials are already undergoing with different technologies such as photodynamic therapy, microwave ablation, radiofrequency ablation, laser, and cryoablation.
So to conclude, we've gone over what the solitary lung nodule is, how to risk stratify a patient to plan what best next step is. And we've also talked about navigation bronchoscopy. Basically, how-- what is it that we're using nowadays to get to the nodule and get a confident diagnosis?
We've gone over electromagnetic navigation, robotic platforms, also talked about using adjuncts with this platform such as the radial endobronchial ultrasound, [INAUDIBLE] cone beam CT. And we mentioned the therapeutic implications.
And with that, I finish my presentation. Thank you for your attention.
DR. MARK DYLEWSKI: Thank you very much, Dr. Sosa. It was an excellent review. Our next speaker is Dr. Raul Valor. He's an interventional and clinical pulmonologist, the Chief of the Department of Medicine who's going to be talking about the use of therapeutic bronchoscopy for the treatment of lung cancer. Thank you very much, and welcome Dr. Valor.
DR. RAUL VALOR: Good evening. My name is Raul Valor, and I'm an interventional and clinical pulmonologist at Baptist. And I'm going to be talking about therapeutic bronchoscopy in the treatment of lung cancer.
I have no conflicts of interest. And at the center of my talk is narrowband imaging, as this modality of evaluating the airways allows us to find lesions that we would otherwise not be able to find.
The way [AUDIO OUT] use a special filter in the regular bronchoscope that filters out certain wavelengths in the white light. And by doing that, it enhances certain vascular structures within the airway.
As we can see here, these are abnormal loops of vascularity that are definitely abnormal.
What I do is that I pretty much do narrow band imaging, and almost all my [INAUDIBLE]. And it's certainly indicated in the patients that have a prior history of aerodigestive squamous cell carcinoma and patients that have a greater than 30 pack year history of smoking.
I'd like to present briefly a case of a 72-year-old patient with history of colon cancer with a 50 pack year history of smoking, and an abnormal CT of the chest referred to me for bronchoscopy for tissue diagnosis of that mass in the lung. It was uncertain whether this was a metastatic lesion.
As we can see there, the patient's lung function is very poor, consistent with advanced COPD. We can see there is a lesion in the right upper lobe here. And the reason I'm showing this is to illustrate further that there is a right upper lobe lesion here. And I'm showing this with the special interest of showing this area here.
That although there is an area of bronchiectasis, there's no mass, per se. During bronchoscopy, during a navigation bronchoscopy to evaluate that [INAUDIBLE] mass, I found this lesion in the secondary carina. When I looked at it closer with narrowband imaging, which I mentioned earlier, it had disappearance.
And when I biopsied it, it yielded squamous cell carcinoma. This is the lesion after it was treated with photodynamic therapy. And after debridement-- and I can tell you that after approximately 18 months of follow up and repeated biopsies in different areas of this lesion after treatment, it was resulted in a complete cure.
The right upper lobe lesion, just out of interest, did show adenocarcinoma. It was a second primary. It was not a metastatic lesion. But this patient could not have had a lobectomy because of her compromised lung function. And especially in light of the fact that there was two concurrent lesions.
So I chose to treat this lesion the [AUDIO OUT] in the secondary carina and the left upper lobe region, the photodynamic therapy, and seems to have been curative.
Jumping right into photodynamic therapy as an overview. Photodynamic therapy uses a photosensitizing agent that's administered intravenously. And there's a uptake by the tissues, and finally clearance. There's selective retention in tumors because of the vascular anatomy of the tumors.
And after 48 hours, a non-thermal light laser is applied, which causes immediate release of singlet oxygen, which in turn causes tumor tissue destruction. In a nutshell, this is how it works. The effects of the singlet oxygen on the mitochondria inactivates the membrane bound enzymes, causing the polarization and the initiation of apoptosis or cell death.
In the nucleus, [INAUDIBLE] fragmentation. And in the membrane, causes lipid peroxidation. Also causes enzyme leakage at the lysosomes. It's indicated in the treatment of microvascular carcinoma in situ of non-small cell lung cancer. And this is detected by screening, as I alluded to earlier. By bronchoscopy and utilizing narrowband imaging is very helpful.
This is particularly done in patients that are either poor surgical candidates or patients that outright refuse surgery. And also in application in obstructive lesions where there's partial or full obstruction of the airway. And it's recommended by the NCCN for treatment of endobronchial obstruction, as we see here in this stage of treatment.
And these are the laser applicators that we utilize. There is a variety of lengths, and depending on whether we're using a rigid or flexible, whether we're placing it within the airway or introducing it within the lesion. And over here on the right, we have the laser generator where you remember this is a non-thermal light laser.
So we utilize up to 200 joules per centimeter. It takes about 15 to 20 minutes to apply the laser to the lesion.
I'd like to show you this other case of a 61-year-old female with a history of breast cancer who had been treated [INAUDIBLE] multiple therapies. Also had a 40 pack year history of smoking. Presented with hemoptysis and underwent bronchoscopy by a colleague, a pulmonologist, who then referred the patient to me.
The patient had advanced COPD, as we can see her lung function there. And imaging showed this right lower lobe mass. Bronchoscopically, the patient had this large obstructing lesion to the right lower lobe, which to anybody who is a bronchoscopist, would appear as a carcinoid tumor, glistening and very sharp margins.
And we're usually taught at bronchoscopy school not to biopsy these because of their tendency to bleed. I went ahead and biopsied it, and it showed a rather rare inflammatory tumor, not typical at all.
And I went on to do bronchoscopy in this patient with photodynamic therapy. The patient received the photosensitive tasting drug. Here's a picture of the application of the laser. And this is immediately after the laser was applied. As you can see, the tumor is much redder and beefier and has shrunk significantly.
And this is after the area was debrided, and you can see that the airway was fully reestablished. These are side by side CTs of before and after.
And the way I do the procedure, patient selection is really paramount. We have to review the imaging very closely and make sure that there is no vascular structures within 4 millimeters of the-- as the penetration is-- could be up to four to five millimeters.
And the last thing we would want is for the effects of the photodynamic therapy to go into one of the vessels, as this would be-- have devastating effects. Patients are consented and medication is injected, usually on a Friday because of the timing. It seems to work out the best.
Patient undergoes bronchoscopy with general anesthesia, and I typically use a LMA airway. The laser application is usually done on a Monday and debridement is then done on Wednesday, possibly Friday, and possibly Monday depending on the amount of tumor burden that's left.
I've gone to do cases that I've done it for several weeks. The effects of the [INAUDIBLE] last for about eight months, but I will continue to do bronchoscopy periodically every three months. Some of the data that has shown the effects of photodynamic therapy in patients with non-small cell lung cancer.
This very ambitious and large cohort compared the effects of photodynamic therapy with radiation alone, radiation and photodynamic therapy, and radiation with chemotherapy. And we can see here some of the results. And their conclusion was that adding endoluminal ablation, such as photodynamic therapy, to other treatments resulted in a significant advantage in survival in patients with non-small cell lung cancer.
Further, a meta analysis done by [INAUDIBLE] in 2003 with 636 patients, and that advanced lung cancer showed a symptomatic relief in virtually all of these patients. And a randomized, controlled trial of-- compared to nd-YAG laser showed that the survival and response in patients with full dynamic therapy was much superior.
PDT has been evaluated directly with checkpoint agents, and it does have a potential immunologic synergism. There's evidence to suggest that photodynamic therapy increases the immunogenicity of dead tumor cells by exposing or creating new antigens and by inducing heat shock proteins that increase the efficacy of antigen [INAUDIBLE].
For more effective tumor specific cytotoxic T cells. And photodynamic therapy can produce tumor cures and long lasting tumor specific immunity, such as memory, as has been shown by studies done in mice.
Now I'd like to talk about spray cryotherapy. And this modality is primarily used in the central airway lesions only. We use liquid nitrogen spray for each five second application. The amount of gas expense to five liters. Therefore, a passive venting technique needs to be applied or else the amount of pressure build up in the airway is such that it results in pnemomediastinum and pneumothorax with possibly devastating consequences.
So essentially, the procedure is done with the patient under general anesthesia. Only central airway tumors are treated because of the passive venting issue. And at the time of spray, the endotracheal tube cuff is deflated, the patient's mouth is opened widely.
Therefore, there is a-- the gas has the ability to egress. And I'll show you this case where there's atelectasis of this lung, and the airway on the left main stem bronchus was completely occluded by tumor. And this was a metastatic thyroid tumor and treated with cryo spray.
This is during the actual procedure of spraying, and this is following debridement. As we can see, there was a significant re-expansion there. Now cryo probe is a different technology and different modality. We use C02 touch probe, and this may be used in more distal lesions in the airway.
And it's very useful also to do debridement, and we even use it in patients with benign process. And the concept is that there is a sudden transfer of gas from a higher to a lower pressure, which leads to a very significant drop in temperature. And we repeatedly freeze and thaw the tissue, which leads to necrosis and crystallization.
And we can see here the process of freezing and thawing these tissues. There are tissues that are sensitive to cryo and there's tissues that are not. Tissues that are sensitive are listed here, and tissues that are resistant are listed here. I'm going to move on in the interest of time.
And I'd like to show you this case of a patient that had atelectasis of this segment of the left upper lobe that had this obstructing lesion in the left main stem bronchus. And this is how it appeared following cryo. This was a benign process, a patient that had aspirated a foreign body and had all this granulation tissue growing around it.
And after some debridement, I was able to retrieve this foreign body that was actually a piece of straw that was crushed and inhaled accidentally. And this is the appearance after the foreign body was removed and the tissue was debrided. APC, or Argon Plasma Coagulation, another modality that we have to treat airway tumors.
It's a non-contact mode. Argon is a gas that conducts electricity. And essentially the way this works, it sprays Argon gas and it has a penetration of three millimeters and it causes superficial coagulation of tissues, as you can see here. We see here examples of Argon plasma coagulation being sprayed, either in a rectilinear or a right angle approach.
The probe is flexible, and the power settings are anywhere between 30 and 60 watts with a flow of 0.3 to 2 liters per minute. We usually apply for two to three seconds at a time and allow the tissue to cool down. The tip needs to be extended well beyond the tip of the bronchoscope, and the target should be kept at about a centimeter of distance.
And that is it for me tonight. Thank you very much for attending.
DR. MARK DYLEWSKI: Thank you very much, Dr. Valor. Very good summary of a complicated subject. Our next speaker is Dr. Paul Kaywin. He's the head of thoracic medical oncology at the Miami Cancer Institute. He will be reviewing treatment of advanced non-small cell lung carcinoma with chemotherapy and the role of immunotherapy. Thank you very much, Dr. Kaywin.
DR. PAUL KAYWIN: Thank you for allowing me to give a presentation on the treatment of advanced non-small cell lung cancer with chemotherapy and the role of immunotherapy. Please note that on this talk, I'm not going to be talking about small cell lung cancer and I'm not going to be going into detail about the treatment of mutated lung cancer with targeted treatments.
I have no disclosures.
So in terms of background, we're going to be concentrating on adenocarcinoma, squamous cell carcinoma. And please note that both of these are smoking related cancers, so cancer prevention, smoking prevention, is always important. Another important item is that before we talk at all about deciding on chemotherapy or immunotherapy is that the standard of care now is to test all lung cancers for mutations.
These are somatic mutations, not inherited germline mutations that can be tested on tumor biopsies, or since cancers can shed their DNA in the blood, we can measure this by liquid biopsy. And as this pie chart shows, we have a number of different mutations that can be detected.
And the purpose of this is because many of these mutations are actionable, where we have treatments that can block these driver mutations. So this needs to be tested before we even consider chemotherapy or immunotherapy.
Now let's take a look at the timeline of the treatments that we're talking about today are fairly modern. Chemotherapy advent in the 1940s, targeted therapies coming into play in the 1980s, and the immune therapy checkpoint inhibitors that we'll talk about coming into play in the 2010s.
If we look at specifically chemotherapy and the timeline of the development of chemotherapy, I just want to point out a few items. Number one, since the 1980s, cisplatin, or carboplatin, platinum doublets are the standard of care. There's never been any studies showing that adding three or four drugs are better than doublets, and we still use those today.
Another highlight is the development of pemetrexed as a medication to treat non-squamous adenocarcinomas. And a third development of importance is the addition of VEGF inhibitors, Vascular Endothelial Growth Factor inhibitors, such as bevacizumab or Avastin, which work in a way to block the microcirculation to the cancer can be added to chemotherapy regimens.
If we look at the timeline for immuno-oncology, the Holy Grail of trying to harness the body's immune system to fight cancer is not new. And even back in the 1890s, Coley noted that in patients with very advanced cancers, if they had life threatening infections with high fevers, that some of their cancers would regress.
And he developed what was called a Coley toxin to try and replicate that. He didn't understand at that time that this had to do with the immune system. That wasn't really understood until the 1900s where immunity having to do with cancer was learned about.
And if we look more currently, it wasn't really until 2011 that the FDA approved the first checkpoint inhibitor, which we'll talk about for melanoma. And now in 2015, the FDA approved treatments for lung cancer with immunotherapies.
So let's talk a little bit about chemotherapy. Chemotherapy works having to do with blocking, cell replication during the cell cycle. Different chemotherapy medicines work at different parts of the cell cycle. Platinum compounds are cell cycle independent and can work in the G1 phase.
Pemetrexed and anti-metabolite block cell cycle replication in the S phase. Topoisomerase is an enzyme that helps to uncoil the DNA helix and can be inhibited by agents such as etoposide in the S and G2 phase. And then the taxanes work as microtubular inhibitors.
The microtubules are what carry the chromosomes to each side of the cell during cell replication.
And chemotherapy, as we know, can have many side effects. Just to highlight a few, bone marrow suppression causing low blood counts, risk of fever, infection, or bleeding. There can be nausea and vomiting, sexual dysfunction, and infertility. Effects on the kidneys, hair loss, to name a few.
Now in distinction, immunotherapy works much differently. Cytotoxic T cells are what our body uses to identify and kill tumor cells. So in this cartoon, we see T cells. And T cells have checkpoints to keep the T cells in balance normally in our body. These checkpoints are protein receptors on the surface of the T cell.
The ones that we are-- look most commonly at have to do with programmed death one, TD-1 antigen on the surface of the T cell, and CTLA-4. Now cancer cells have ligands, the PDL-1 ligand, that can block the PD-1. And by doing that, can inhibit T cells.
So tumor cells have a way of becoming stealth downregulating T cells so that they're not recognized by our own cytotoxic T cells. Tumor cells, as depicted in this cartoon by these blue dots, also have antigens that can be picked up by antigen presenting cells that also interact with our T cells.
And there is an antigen, surface antigen, on the T cell, surface protein on the T cell. CTLA-4, which is an inhibitory protein. So now we have agents that can activate our T cells and unleash a killing of tumor cells. For example, we have PD-1 antibodies that have been developed, such as nivolumab and pembrolizumab.
By blocking the PD-1 receptor, it prevents this connection between the tumor cell, PDL-1, and PD-1 from downregulating and inhibiting the T cell. So the T cell becomes activated. We have PDL-1 blockers such as atezolizumab or [INAUDIBLE] or durvalumab, which block the PDL-1 receptor on the tumor cell and accomplish the same thing by preventing this tumor cell from inhibiting the T cell.
CTLA-4 is inhibitory and ipilimumab is an antibody that unblocks CTLA-4. And by unblocking CTLA-4, the inhibitory effects of CTLA-4 are decreased and thus activating T cells. So if we look at side effects of immunotherapy, these are quite different than chemotherapy.
Immunotherapy can cause side effects by overstimulating the body's T cells, and can cause inflammation or many types of itis, if you will, in different organs of the body. There can be skin rash or dermatitis. There can be colitis or diarrhea. There can be pneumonitis.
You can develop thyroiditis, uveitis, nephritis, hepatitis. Fortunately, these are uncommon side effects from immunotherapy. If they do occur, they can be treated either by holding the immunotherapy or using immunosuppressants to cool down the overstimulated immune system, for example, with steroids.
Now how do we make decisions, then? We have chemotherapy, we have immunotherapy. Both on the chemotherapy side, decisions of what medications to use are based on histology. Squamous cell carcinoma, we tend to use taxanes and platinum. Remember the doublets?
In adenocarcinoma, we use pemetrexed and platinum in that doublet. There are other regimens as well, but those are the most common. How do we make decisions about immunotherapy? Well, on the tumor, we can now measure the PDL-1 as a biomarker for immunotherapy.
And these slides depict an adenocarcinoma. And the brown on these slides represent staining of the PDL-1 on the cancer. And there's different degrees of staining, and the pathologist can then score the actual percent of tumor cells that are PDL-1 positive and give us a report where the tumor can be rated, or scored, as having a low or an intermediate or a high PDL-1 score.
The higher the PDL-1 score, the more likely the tumor is to respond to immunotherapy.
So let's look, then, at how we make decisions. Should we be using immunotherapy or chemotherapy or both? And there have been many, many clinical trials done. Primarily, the clinical trials used to get FDA approval for immunotherapies that have shown the benefit of immunotherapy compared to chemotherapy.
This is a meta analysis showing a forest plot, where anything to the left favors immunotherapy versus chemotherapy. So in this meta analysis of many studies that were done, we know that immunotherapy, especially in the right population of patients, especially people that have high PDL-1s have had better outcomes in terms of overall survival compared to chemotherapy.
And in this meta analysis, we can see that responses to immunotherapy do vary based on the level of PDL-1. Here we have PDL-1. Less than 1%, an intermediate group of 1 to 49%, and a high group. And we can see that the higher the PDL-1 in this forest plot, to the right would favor immunotherapy responses.
So the higher the PDL-1, the more likely there would be a response to immunotherapy. Now how do we look at the combination if we use immunotherapy alone or immunotherapy combined with chemotherapy? In this meta analysis, we can see that regardless of the PDL-1 level-- whether it's low, intermediate, or high-- in this forest plot, to the left would favor immunotherapy combined with chemotherapy.
So we know that if we combine immunotherapy with chemotherapy, there can be better outcomes. So we need to then think about the treatment decisions when faced with a stage four patient with lung cancer. And there are a number of important decision points that have to be thought out.
First and foremost, we have to establish the patient's overall performance status and urgency to treat. It can often take one to two weeks to get results of the molecular and PDL-1 testing. So if someone needs urgent treatment, we would first treat with chemotherapy. Mutated cancers, or even high PDL-1 cancers, will still respond to chemotherapy.
So if someone's in the hospital, is very sick and we can't wait for these tests, we would start with chemotherapy.
The next decision point is, as I've mentioned before, we must establish if there are any actionable mutations present to enable treatment with an oral targeted biologic. An example of this would be using osimertinib to treat an EGFR mutation. Beyond that, if we are not going to be using a biologic, we then need to establish whether or not the patient has any autoimmune contraindications to immunotherapy.
If someone has active rheumatoid arthritis or very active inflammatory bowel disease, immunotherapy can exacerbate those autoimmune problems and immunotherapy may be contraindicated.
Once we decide that, yes, someone is a candidate for immunotherapy, we then have a number of options and we tailor the decision making to each individual patient. We have an option for single agent immunotherapy, especially if there's a high PDL-1 score or if chemotherapy is either refused or would not be tolerated.
There are regimens combining single agent immunotherapy with chemotherapy, and sometimes we can also add a VEGF inhibitor such as bevacizumab, as was shown in this Empower 150 study. We have additional option to use non-chemotherapy dual immunotherapy with a checkpoint inhibitor, the CTLA-4 inhibitor ipilimumab and a PD-1 inhibitor nivolumab, as was demonstrated in the CheckMate 227 clinical trial.
We have to be cautious with this regimen because there is the potential for more immune related adverse event side effects. And then finally, we have options to use a brief course of chemotherapy since responses to chemotherapy may occur faster than immunotherapy.
So there are regimens, as was demonstrated in the Checkpoint 9 LA study, to use brief two cycles of chemotherapy and immunotherapy to begin with, and then dropping the chemotherapy and continuing the immunotherapy.
So we have a number of different options, as demonstrated on this slide, where we can see for both squamous and non-squamous cell cancer, based on the level of PDL-1, this demonstrates the key clinical trials that have led to approval of a number of different combinations of immunotherapy alone, immunotherapy with-- combined with chemotherapy for the treatment of non-small cell lung cancer.
And we tailor the treatment to each individual patient's particular situation.
Just to give an idea, then, of how successful we are-- just to finish up-- I want to present the Keynote 189 data, which was one of the first trials using pembrolizumab, a PD-1 inhibitor, and a platinum doublet compared to a platinum doublet alone. So that was the randomized trial.
And we now have data going out-- going on four years that show, for example, for people with high PDL-1 scores of greater than 50%, we now have survival-- 50% survivals going out over 36 months in a population of patients where historically, with chemotherapy alone, the survival may be less than one year.
Not everyone has a high PDL-1 score, and the results for-- are not as favorable in people who have lower PDL-1 scores. But this is the state of the art, where we have quite a bit of improved survival with immunotherapy. I thank you for allowing me to give this presentation.
DR. MARK DYLEWSKI: Thank you very much, Dr. Kaywin. Excellent summary of a very complex topic. Our next speaker is from the Lynn Cancer Center. It's Matthew-- Dr. Mathews. He's head of hematology and oncology at Boca Regional Medical Center. He's going to give a talk on treatment of advanced non-small cell lung cancer with targeted therapies.
Welcome, Dr. Mathews. I thank you for speaking here today.
DR. MATTHEN MATTHEW: I am Matthen Mathew. I'm one of the medical oncologists at the Lynn Cancer Institute, and I'm going to talk today about the treatment of advanced non-small cell lung cancer with a particular focus on targeted therapies.
So I don't have any disclosures, and I won't be discussing any unlabeled or unapproved use of any drugs.
So to quickly go over some of the learning objectives, my goal today is to talk-- review the molecular alterations that we know about in non-small cell lung cancer and also go over the current landscape of targeted therapies in our treatment. I'm also going to quickly describe some of the issues we have with biomarker testing, and also the importance of biomarker testing with regards to understanding more about the genomic alterations.
So just some background, we've really moved away from the understanding of non-small cell lung cancer as one disease and we really started to subdivide it into a group of both histologically defined, as well as molecularly defined, diseases. Initially in-- particular with regards to squamous cell versus adenocarcinomas.
But now, really, much more of a molecular, granular understanding of these tumors has helped us define and refine therapies for them. And what we can see here is also this pie chart that shows the box of unknowns has really gotten slimmer and slimmer. And here's where we stand in 2021.
And this is-- on the pie chart on the left is actually looking at molecular alterations seen in patients with adenocarcinomas. And these are tumors that we oftentimes see in patients who are never smokers, or light smokers. Also women as well as younger patients. And there's a number of different alterations, not just single nucleotide variants.
But also fusion events, as well as copy number alterations that we can see in these tumors. And I'm going to briefly review some of them as well as the targeted therapies that we now have associated with them. But I'm not going to have time to go through all of them in detail, though there is an ever changing landscape on these mutations as well as the targeted therapy associated with that.
So to start with EGFR, this is the first molecular alteration identified in lung cancer. It was identified back in 2004, and some of the initial inhibitors for this actually, we didn't understand that EGFR was the reason that patients were doing so well. It was only when they went back to the data and looked at it did they find that those who are enriched with EGFR mutations actually did well with the therapies.
These-- so EGFR is a member of the Herr family. It's actually Herr-3, and receptor ligand binding which really leads to receptor differentiation downstream signaling and cellular proliferation. The spectrum of these mutations is generally focused on the tyrosine kinase domain, and the majority of these mutations are actually either in exon 19 with a deletion event or a point mutation in exon 21 known as L858.
Together, these comprise about 80% to 90% of the mutations in EGFR. And importantly, these are sensitive to tyrosine kinase inhibitors. There are non-canonical mutations as well that we see in exon 20. And importantly, this is recent now, therapy for this as well as exon 18 and other mutations.
But we're going to focus mostly on the sensitive sensitizing mutations in exon 19 and 21. Now drug therapy has advanced from first generation and tyrosine kinase inhibitors to secondary inhibitors like erlotinib. And now, the third generation inhibitors like osimertinib or Tagrisso.
And this is data from the FLAURA study, which was looking at first line osimertinib in metastatic non-small cell lung cancer harboring a sensitizing EGFR mutation. And on the left, we see the waterfall plot with this drug, and it's really quite impressive, with an objective response rate of 80%.
And on the right here, in the Kaplan-Meier curve, we're seeing median overall survivals ranging now in the order of 30 over three years. And that's compared to the older generation TKIs.
This actually allowed osimertinib to be moved towards the non-metastatic setting, and this is data from the ADAURA study looking at osimertinib compared to placebo in patients with stage 1B to 3A non-small cell lung cancer harboring an EGFR mutation that had received surgery and adjuvant chemotherapy, and subsequently got either osimertinib for three years or placebo.
And what we can see here is a really impressive separation of the curves between Tagrisso and the control arm. This led to a hazard ratio of 0.17. That's closer to a P value than a hazard ratio. And a markedly improved disease free survival and let's the approval of osimertinib in this adjuvant setting.
And this benefit was actually seen across subgroups that we can see here in the forest plot on the right.
So sensitizing EGFR mutations, the most common actual alteration that we see in lung cancer-- non-small cell lung cancer and third generation TKI therapy with osimertinib is now FDA approved both for the metastatic as well as the non-metastatic or adjuvant setting. And these drugs are generally very manageable and well tolerated.
Toxicity profile that we do see involved. Sometimes the wild type EGFR in the skin GI, also cardiovascular side effects. But for the most part, again, these are manageable.
Moving on to ALK alterations in non-small cell lung cancer. Now importantly, there are-- these are about 3% to 5% of patients with non-small cell lung cancer with a driver mutation. Importantly there are mutations in tyrosine kinase domain, like we saw with EGFR. But for the most part we care about with ALK is actually a fusion event.
And this is when the kinase domain is attached to five prime partner for a different gene. And most commonly in non-small cell lung cancer, we see ML-4. And again, across the board, fusion events in non-small cell lung cancer, these are what the TKIs are approved for. We do not have approvals at this point in time for ALK viewpoint mutations. And that's an important principle to understand.
With regards to the tyrosine kinase inhibitors, we have older generation tyrosine kinase inhibitors like crizotinib and also newer generation TKI such as lorlatinib. And these are some of the hazard ratio-- these are some of the Kaplan-Meier curves and some of the data for the second and third generation TKIs here.
And you can see excellent separation of curves. These are all compared to crizotinib in the [INAUDIBLE] arm, with hazard ratios ranging from 0.228 and with [INAUDIBLE] and all the way up to 0.51. So again, excellent data for the newer generation TKIs. It just also brings up a point here with these alterations that we do unfortunately see resistance invariably, both with almost all of the targeted mutations and targeted therapies that we find.
And these resistance patterns can really be variable. So we can see target alterations, for example, if we have changes in the tyrosine kinase domain, point mutations that make it insensitive to inhibitors. That can be a problem. And also we can see bypass track signaling. For example, with BRAF mutations.
Initially when we targeted BRAF alone, we found that there was upstream signaling through the MEK pathway. And so now we do BRAF and MEK combination therapies to avoid that. And there are also other pathways such as efflux pumps and histologic transformation like small cell transformation EGFR, which I'm not going to go into.
With ALK resistance, it's actually a fairly heterogeneous resistance pattern that we see compared to EGFR which has a more-- relatively small number of resistance mutations. The important thing to understand here is regardless of the second generation TKI, over 50% of patients are going to develop resistance.
One of the more common resistance mutations is G12-O12-R. And I think it's actually important to note that the newer generation TKIs, particularly lorlatinib, these are IC50 curves, or plots, are actually very effective against these mutations. But oftentimes, it does end up becoming a game of cat and mouse, and we're always trying to target the resistance mechanisms that these alterations can-- these tumors can develop.
So with ALK, I think that it's important to understand how these various TKIs can be sequenced in the ongoing studies to understand how to best use them. Currently alectinib and brigatinib and lorlatinib are all approved for the front line setting. There are heterogenous resistance mechanisms.
And important with regards to ALK is that CNS, the Central Nervous System, can be a sanctuary site and should be monitored very closely.
Moving on to RET fusions. These are about 2% of non-small cell lung cancer with a driver mutation. And there's really two different, again, types of alterations. We can see there are nonsynonymous point mutations in the tyrosine kinase domain. With these, we actually see these more with thyroid cancer, particularly multiple endocrine neoplasia.
But again, for lung cancer what we really care about is the fusion events. And this is when the three prime RET tyrosine kinase domain is attached to five prime partner, often CCDC6 or [INAUDIBLE] 4 in non-small cell lung cancer.
I think RET inhibition is a great example of how rational drug design has led to improved targeted therapies. We used to have multi kinase inhibitors like cabozantinib that you can see on the kinome plot here, had a fairly broad target inhibition. Whereas the more selective inhibitors now, [INAUDIBLE] itself or [INAUDIBLE], are much more effective at targeting RET specifically and much less side effects than we saw with the multi kinase inhibitors.
And this is data from phase two studies looking at [INAUDIBLE] as well as [INAUDIBLE] and excellent waterfall plots objective response rates ranging from 60% to 80% here in treatment naive patients. And this led to the FDA approval for both of these agents in RET fusion, as well as RET mutated thyroid cancer.
RET fusion lung cancer as well as RET mutated thyroid cancer. And there are-- these are both tolerated very well overall. We do see some dry mouth transaminitis and some GI side effects, but for the most part, much better tolerated than chemotherapy. And again, substantially improved response rates and durable disease control here.
I'm going to quickly go through KRAS. Again, this used to be thought of as an undruggable target. Particularly, it's not a tyrosine kinase, it's a GT paste. But now with an understanding of some of the more-- the specific KRAS mutations, particularly G12C, we found that we can target it by inactivating KRAS by combining these drugs with the inactivated GDP found-- form of KRAS.
And so two of the drugs here, sotorasib in the CodeBreak study and adagrasib in the [INAUDIBLE] study, we see excellent objective response rates in the refractory setting. So essentially second line 32 to 45% response rates. And this actually led to the FDA approval of sotorasib with KRAS G12C non-small cell lung cancer in the second line setting.
So this is a timeline of the approvals that we've seen recently. And again, if you look at the blue boxes, these are the targeted therapies and very impressive in the last couple of years. We've had an explosion of targeted therapies that have been approved. Just looking at some of the mutations here, we have over 20 drugs that are now approved for over 10 different molecular targets.
Most recently here, EGF exon 20. These were actually approved in September of 2021. So a lot of options that we have at our disposal now. One of the challenges that I find is really that biomarker testing can be an issue, there's a lot of different names that we have to be aware of. Just fusion events, RNA sequencing, NGS. It can become somewhat of an alphabet soup.
So keeping track of this and talking to molecular pathologists can be helpful. There's also a very crowded space with regards to testing. There are a number of companies that are doing testing these days. Also how to interpret the results can be a challenge. Again, as I talked about, some of the alterations are very important to understand because if you see, for example, with NTRK, if you see a mutation event, that is actually not targetable or FDA approved.
But if you do see a fusion event, for example the TV 6 and track three fusion, that is targetable. So understand the differences between these are important. And lastly, I think it's been excellent that we now have plasma based or liquid biopsies that we can use to better, which are less invasive for patients and can also give us a better understanding of the global somatic burden-- alteration burden.
One of the challenges is it really requires a certain amount of disease to be able to pick up these somatic alterations. If you have just a small tumor, you might not see a mutation that is actually present. So again, the gold standard here is a tissue specimen, particularly if there's a limited version of disease that we see there.
But I think it's really going to change the landscape moving forward, as we start to use this testing more commonly. And one thing to understand is we really just need to improve our testing rates overall. This is a study presented at ASCO this year that showed in a US Oncology database from 2018 to 2020, there was less than 50% of providers that were ordering testing for all of these five biomarkers.
And that's really a challenge because if we don't order the test, we're not going to find the results that can be used as a drug target. And I think that the-- particularly targeted therapies, I think that the improvement in survival has been really remarkable, ranging from chemotherapy in the old days with overall survivals on the order of eight months.
Immunotherapy that you've heard about in this series as well as has also improved survival on the order of 30% at five years. But really, with drugs such as alectinib and ALK rearranged non-small cell lung cancer, the five year overall survival is remarkable at over 50%.
So just to summarize here, a better understanding of the molecular characteristics of non-small cell lung cancer has really led to novel targeted therapies. There are now over 20 drugs that I mentioned for 10 driver mutations, including seven in the front line setting. But we really got to order the test to make sure we can find these mutations and alterations.
And hopefully, the use of plasma based testing will lead to better testing platforms and more widespread utilization. And there are some very exciting novel targeting approaches, also including antibody drug conjugates as well as pro-tax which are essentially targeting proteins for ubiquitin degradation that are on the horizon.
So I think that leads us to the-- what I really believe is the promise of precision medicine here. I think our goal is really to be able to talk-- to find a particular molecular mutation or alteration for each particular patient's tumor, and be able to match up therapy for that alteration.
And sometimes, also at resistant-- when we find resistance patterns, to go back and do the biomarker testing to look for if there's a particular resistance mutation that we can further target. And the other-- on the right hand side here, the other thing which I think is remarkable is how there's been explosion of biomarker patient support groups.
I was recently part of a lung cancer support group here at Lynn Cancer, and there was a patient that had an ALK rearrangement who was part of a ALK support group. And he just mentioned how great it had been to be able to talk to other patients dealing with these alterations, both from a research platform to find novel research strategies, as well to talk about side effects of therapy.
So I think it's really great, and I do encourage patients to sign up for this. And with that, I'll stop. And thank you very much for your time.
DR. MARK DYLEWSKI: Thank you, Dr. Mathews. It was an excellent overview of the molecular characteristics of non-small cell carcinoma. Thank you very much for speaking here today. Our next speaker, and our last speaker. And I'd just like to remind the audience that there is a Q&A button at the bottom if there's any questions that you want to type in for our esteemed speakers to answer.
Please use that button. But our last speaker is Dr. Federico Albrecht, a close colleague of mine who's a thoracic oncologist at the Miami Cancer Institute. And he'll do an update on the role of neoadjuvant therapy, defining the role of immunotherapy in surgically susceptible lung cancer patients. Thank you, Dr. Albrecht.
DR. FEDERICO ALBRECHT: My name is Federico Albrecht. I'm one of the thoracic medical oncologists at the Miami Cancer Institute. I will present this evening an update in the role of neoadjuvant therapy. Defining the role of immunotherapy for the surgically detectable lung cancer patients.
These are my disclosures.
Well, it is very likely that in the near future, we'll have an FDA approved option to use immunotherapy in combination to chemotherapy as a neoadjuvant treatment option in resectable lung cancer patients. So what are the potential advantages of a neoadjuvant therapy approach?
Well, for one, it is the improved patient compliance with therapy. There is also evidence to suggest that a potential for improved immune activation with neoadjuvant compared to adjuvant approach. As an example, what I'm trying to say is there was a trial for actually patients with melanoma.
Patients were treated with neoadjuvant immunotherapy. And those patients had a greater expansion of tumor infiltrating T cells in the peripheral blood, as well as expansion of new T cells clones compared to patients that received immunotherapy in an adjuvant form.
Suggesting that there is something very important about the intact tumor burden in eliciting a robust immune tumor response. In addition, pathologic response can serve as a potential surrogate for clinical efficacy. And a more extensive resection tissue allows for better and in-depth analysis.
So if we identify an early efficacy endpoint as a surrogate market for a long-term benefit, that could lead to early approvals in the perioperative setting for early diagnosed lung cancer patients. Pathologic response is already being utilized as a surrogate marker in breast cancer.
There is data, as you can see here, that a major pathologic response after neoadjuvant chemotherapy-- which is defined by less than or equal to 10% tumor cells in a resected specimen-- is associated with a survival benefit in patients with breast cancer. And the association seems to be even stronger than what is seen by radiologic [INAUDIBLE] criteria.
So could pathologic response be used in lung cancer patients as well? We will see some data coming up.
So what are the potential disadvantages of a neoadjuvant immunotherapy or chemotherapy approach? One is that neoadjuvant therapy is not routinely or commonly used as is adjuvant chemotherapy in current lung cancer clinical practice. Other proposed disadvantage is that the tumor might progress on neoadjuvant therapy, preventing a potential curative surgery.
There is a perception that the therapeutic effect of immunotherapy can make surgery more complicated or more dangerous. So what data do we have?
There have been multiple early phase trials studying the safety and efficacy of immunotherapy plus or minus CTLA-4, as you can see here in this table. The largest of these studies is the LCMC-3. There has been newer data of this study presented at the recent World Land Conference.
This is the design of the LCM-3 study. It is a single arm phase two trial in which patients with respectable untreated lung cancer with stages 1B to 3A receive two cycles of atezolizumab followed by surgical resection and then standard of care. The study included 181 patients, and the primary endpoint of the study was major pathologic response.
The secondary endpoints + pathologic response, radiologic response, and the exploratory endpoints were overall survival. This is free survival biomarkers and safety.
So what are the results of the study? As you can see, 88% of patients were able to proceed to surgery, of whom 92% achieved an R0 resection and 43% had a pathologic downstaging, suggesting that neoadjuvant immunotherapy did not impede surgical outcomes.
Second, a major pathologic response was seen in 21% of patients, seen here in blue. And a complete pathologic response of 7%, seen here in orange. Meaning no tumor was seen in the resected-- in the resected specimen.
As a median follow up of 2.1 year, the median disease free survival and overall survival have not been yet reached. We see encouraging signs of clinical efficacy when the results overlaid over historical data seems to be better with neoadjuvant immunotherapy, both for stages one and two, in blue, and stages three in red.
So this data is in reference to neoadjuvant immunotherapy alone. So what about the combination of immunotherapy plus chemotherapy used as neoadjuvant treatment? There are several ongoing phase two trials that have reported actually so far.
And these trials have been very impressive, with the majority of patients going on to receive surgical resection, showing impressive pathologic responses. When you combine this data, the major pathologic response is in the order of around 58%, with a pathologic complete response in the order of 33%.
Remember that pathologic complete response is when there is no viable tumor in the removed specimen.
So what this suggests is that a pathologic complete response is potentially a viable surrogate marker. So pathologic responses have been used as an endpoint in several of these ongoing phase three neoadjuvant studies, as you can see here in this table.
We have early data of one of these studies-- actually, it's probably the first one, the CheckMate 816, that has been presented at our last ASCO conference this year. So this is the design of the phase three CheckMate 816 study, which included newly diagnosed respectable lung cancer patients with stages 1B to 3A, with no evidence of EGFR or ALK mutations.
The patients were stratified by stage, PDL-1, and gender.
So we had about 58 patients were randomized to receive neoadjuvant nivolumab plus chemotherapy for three cycles versus neoadjuvant chemotherapy alone for three cycles, followed by surgery. Within six weeks, both treatment and then optional adjuvant chemotherapy plus/minus radiation therapy, as per standard of care.
The principal endpoint of this study were pathologic complete response, and eventually survival. Secondary endpoints were major pathologic response, overall survival, time to death, or distant metastases. And finally, exploratory endpoints were overall response rate and predictive biomarkers, including PDL-1, TMB, and circulating DNA.
Looking at the baseline characteristics, populations were quite well balanced. And in terms of disease stage, 2/3 of patients in both arms had stage three disease, which is the highest risk of recurrence.
So what about these surgical endpoints? When you look at all the patients, you could see that 94 of-- 94% of patients completed neoadjuvant therapy with nivolumab plus chemotherapy, compared to 85% with chemotherapy alone. And overall, as you can see here, 83% of patients went to have surgery in the nivolumab plus chemotherapy versus 75% in the chemotherapy alone arm.
The medium duration of surgery was shorter in the nivolumab plus chemotherapy versus chemotherapy arm.
Of those patients that had surgery canceled, 7% were because of disease progression in the nivolumab plus chemotherapy arm versus 10% in the chemotherapy arm.
So when you broke down by stage, you can see that it was primarily driving-- driven by the locally advanced stage of three patients, in which 83% received surgery after nivolumab plus chemotherapy compared to 72% of patients with stage three that were treated with chemotherapy neoadjuvant alone.
Patients with stage three had a greater disease progression in the chemotherapy only arm. And again, the duration of surgery was longer, as you can see here, in patients that received chemotherapy alone.
Other key surgical outcomes include the fact that the median time from the last dose of neoadjuvant therapy to surgery was equivalent between both arms, suggesting that neoadjuvant immunotherapy when combined with chemotherapy did not lead to any surgical delay.
In addition, in patients with stage 3A disease treated with chemotherapy alone, there was a greater rate of conversion from minimally invasive surgery to open thoracotomy, and a greater percentage requiring pneumonectomy 30% versus 17%. Hospital stay and surgical related complications were similar between both arms, as were the R0 resection, suggesting that immunotherapy with chemotherapy may actually be better than neoadjuvant chemotherapy alone, allowing safer, faster, and better surgery.
So what about the pathologic endpoints? Remember, the pathologic response complete was the primary endpoint of the study. In the intend-to-treat population, a pathologic complete response was seen in 24% of patients in the nivolumab plus chemotherapy arm compared to 2.2% with chemotherapy alone.
Now, when you look at this subgroup analysis of this study, the pathologic complete response appears to occur independent of disease stage, disease histology, as well as PDL-1 status or tumor mutation burden or choice of platinum chemotherapy.
When we look at the major pathologic response, which again is defined by less than or equal to 10% viable tumor cells, we see an MPR of 36.9% in the group treated with nivolumab plus chemotherapy compared to 8.9% in those treated with neoadjuvant chemotherapy only.
So more importantly, the depth of pathologic response was more pronounced in those treated with immunotherapy plus chemotherapy. The medium viable tumor cells were 10% in the nivolumab plus chemotherapy arm versus 74% in the chemotherapy arm.
Also, radiologic response was higher in the nivolumab plus chemotherapy than in the neoadjuvant chemotherapy alone. In regards to overall response rate and radiologic staging, we see that nivolumab was added to chemotherapy. The overall response rate was increased from 37 to 54%.
Most were partial responses and stable disease, as you can see here. Patients with radiological stages were 31% versus 24%.
So what about safety? Overall, adverse events were similar in both arms. There were similar treatment related adverse events leading to discontinuation of therapy, as well as treatment related serious adverse events. The breakdown here is what you normally expect to see with immunotherapy and chemotherapy.
Adverse events were well balanced between both groups. And when you look specifically to immune mediated side effects, most of these tended to be lower grade, especially when compared to patients receiving immunotherapy in the metastatic setting.
So in summary, what are the benefits of neoadjuvant immunotherapy in respectable lung cancer? Well, the CheckMate 816 showed statistically significant improvement in the primary endpoint of pathologic complete response that could eventually be used as a surrogate marker for future clinical trials and clinical benefit.
As the CheckMate 816 showed that the major pathologic response and the overall response rate were improved when immunotherapy was added to chemotherapy prior surgery. This study continues to mature for the event free survival primary endpoint. We also learned that the addition of immunotherapy to chemotherapy in a neoadjuvant form is durable, safe, and did not impede the feasibility of surgery.
Interesting is to mention that the circulating tumor DNA clearance was more frequent when nivolumab was added to chemotherapy, and appears to be associated with pathologic complete response. The CheckMate 816 is the first phase three study to show the benefit of immunotherapy plus chemotherapy combination for respectable non-small cell lung cancer, and could become a potential new neoadjuvant option for this group of patients.
This is the end of the presentation. For your attention, I thank you very much.
DR. MARK DYLEWSKI: Thank you very much, Dr. Albrecht. It was an excellent summary. I think we can all agree that one common theme with the treatment of lung cancer is that it's a team sport. It involves a multidisciplinary approach of many specialties to achieve the best outcome.
I want to extend my appreciation to all the attendees and all the esteemed speakers today. I just want to say that we'll leave the question and answer icon open for another one or two minutes. If there's any final questions, please send them through the Q&A and we'll-- our esteemed speakers will try our best to answer them as quickly and appropriately as possible.
Otherwise, we'll say good night and thank you, everybody, for attending the Miami Cancer Institute Lung Cancer Symposium. Thank you very much.
Originally Webcast: November 2, 2021 - 6:00 - 8:30 PM EDT
The current state of knowledge regarding multi-modality treatment for lung cancer is continually changing and it is important for surgeons, medical oncologists, radiation oncologists and pulmonologist to stay up-to-date on the latest recommendations for the management of the disease. Dr. Mark Dylewski and the distinguished faculty from Miami Cancer Institute and Lynn Cancer Institute discuss new advances in the management of lung cancer.
Internists, Hospitalists, General Practitioners, Pulmonologists, General Surgeons, Thoracic Surgeons, Obstetricians and Gynecologists, Oncologists, Radiation Oncologists, Nurses, Dietitians, Pharmacists and techs, Respiratory Therapists, Patient Navigators and all other interested healthcare professionals.
Related Videos